WO2003062405A2 - Method for inducing differentiation of embryonic stem cells - Google Patents

Method for inducing differentiation of embryonic stem cells Download PDF

Info

Publication number
WO2003062405A2
WO2003062405A2 PCT/JP2003/000699 JP0300699W WO03062405A2 WO 2003062405 A2 WO2003062405 A2 WO 2003062405A2 JP 0300699 W JP0300699 W JP 0300699W WO 03062405 A2 WO03062405 A2 WO 03062405A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
medium
insulin
cell
patient
Prior art date
Application number
PCT/JP2003/000699
Other languages
French (fr)
Other versions
WO2003062405A3 (en
Inventor
Kazutomo Inoue
Dohoon Kim
Yanjun Gu
Michiyo Ishii
Original Assignee
Yugengaisha Okuma Contactlens Kenkyujo
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Yugengaisha Okuma Contactlens Kenkyujo filed Critical Yugengaisha Okuma Contactlens Kenkyujo
Priority to JP2003562273A priority Critical patent/JP2005514944A/en
Publication of WO2003062405A2 publication Critical patent/WO2003062405A2/en
Publication of WO2003062405A3 publication Critical patent/WO2003062405A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0676Pancreatic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0619Neurons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/46Amines, e.g. putrescine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/235Leukemia inhibitory factor [LIF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • C12N2501/392Sexual steroids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/58Adhesion molecules, e.g. ICAM, VCAM, CD18 (ligand), CD11 (ligand), CD49 (ligand)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells

Definitions

  • the present invention relates to a method for inducing differentiation of mammalian embryonic stem cells into functioning cells.
  • the present invention also relates to the functioning cells obtained by the present invention and a method for treatment of a patient by implanting functioning cells to the patient. ART RELATED
  • Pluripotent stem cells have been derived from two embryonic sources.
  • Embryonic stem (ES) cells are derived from the inner cell mass of preimplantation embryos, and embryonic germ (EG) cells are derived from primordial germ cells (PGCs). Both ES and EG cells are pluripotent and demonstrate germ-line transmission in experimentally produced chimeras.
  • ES and EG cells share several morphological characteristics such as high levels of intracellular alkaline phosphatase (AP), and presentation of specific cell surface glycolipids and glycoproteins. These properties are characteristic of, but not specific for, pluripotent stem cells.
  • AP alkaline phosphatase
  • stem cell is a cell that has the ability to divide (self-replication) for indefinite periods- often throughout the life of the organism. Under the right conditions, or given the right signals, stem cells can give rise (differentiate) to the many different cell types that make up the organism.
  • SDIA stromal cell-derived inducing activity
  • BMP4 acts as an antineuralizing morphogen in Xenopus, suppresses SDIA-induced neuralization and promotes epidermal differentiation.
  • a high proportion of tyrosine hydroxylase-positive neurons producing dopamine are obtained from SDIA-treated ES cells.
  • SDIA-induced dopaminergic neurons integrate into the mouse striatum and remain positive for tyrosine hydroxylase expression.
  • Neural induction by SDIA provides a new powerful tool for both basic neuroscience research and therapeutic applications.
  • mouse embryonic stem cells have been introduced as a new potential source for cell therapy in type 1 diabetic patients (Diabetes 49: 157- 162 (2000), the disclosure of the publication is herein incorporated by reference).
  • Using a cell-trapping system they have obtained an insulin-secreting cell clone from undifferentiated ES cells.
  • the construction used allows the expression of a neomycin selection system under the control of the regulatory regions of the human insulin gene.
  • the chimeric gene also contained a hygromycin resistance gene (pGK-hygro) to select transfected cells.
  • Clusters obtained from this clone were implanted in the spleen of streptozotocin- induced diabetic animals. Hyperglycemia of the transplanted animals were normalized within one week and their body weight were restored in 4 weeks. Whereas slower recovery was observed in the transplanted animals than control mice in an intraperitoneal glucose tolerance test, blood glucose levers after meal load were normalized in a similar manner. This approach opens new possibilities for tissue transplantation in the treatment of typel and type 2 diabetes and offers an alternative to gene therapy.
  • hES pluripotent undifferentiated human embryonic stem cells
  • Su-Chun Zhang et al. disclose in vitro differentiation, enrichment, and transplantation of neural precursor cells from human ES cells.
  • differentiating ES cells formed large numbers of neural tube-like structures in the presence of fibroblast growth factor 2 (FGF-2).
  • FGF-2 fibroblast growth factor 2
  • Neural precursors within these formations were isolated by selective enzymatic digestion and further purified on the basis of differential adhesion. Following withdrawal of FGF-2, they differentiated into neurons, astrocytes, and oligodendrocytes.
  • human ES cell-derived neural precursors were incorporated into a variety of brain regions, where they differentiated into both neurons and astrocytes. No teratoma formation was observed in the transplant recipients. These results depict human ES cells as a source of transplantable neural precursors for possible nervous system repair.
  • Nadya Lumelsky et al. disclose a series of experiments in which they induced mouse embryonic cells to differentiate into insulin-secreting structures that resembled pancreatic islet (Science 292, 1389-1394 (2001), the disclosure of the publication is incorporated herein by reference). They have generated cells expressing insulin and other pancreatic endocrine hormones from mouse ES cells. The cells self-assemble to form three-dimensional cluster similar in topology to normal pancreatic islets where pancreatic cell types are in close association with neurons. Glucose triggers insulin release from these cell clusters by mechanisms similar to those employed in vivo. When injected into diabetic mice, the insulin-producing cells undergo rapid vascularization and maintain a clustered, islet-like organization.
  • the insulin-producing cells obtained by Lumelsky did not express pancreatic specific markers, amylase and carboxypeptidase. Further, Lumelsky grafted the insulin-producing cells into a diabetic model animal but failed to observe a sustained correction of hyperglycemia in the model animal.
  • pancreas transplantation Seven million people in Japan and 16 million people in the United States are affected by type I diabetics. At present, daily insulin administration or allogenic pancreas transplantation is em ployed for treatment of diabetics. Although the overal l success rates of the pancreas transplantation have significantly increased , organ transplantation requires very invasive surgery and life-long immunosurpressive treatments, wh ich significantly strain the patient. Further, availability of donor organs is stil l serious problem preventing the operation to be popular. Therefore, development of a simple and universal treatment for diabetes is desired. SU MMARY OF TH E I NVENTION
  • One object of the present invention is to provide a novel method for inducing differentiation of pluripotent embryonic stem cel ls into functioning cells, especially pancreatic islet like cell clusters and nerve l ike cells. Another object of the present invention to provide a method for treating a patient having disorders in pancreatic islet function.
  • Another object of the present invention is to provide a method for treating a patient having neuronal degeneration or spinal code disorders.
  • Further object of the present i nvention is to provide functioning cells whi ch are derived from mammalian ES cel ls and exhibit pancreatic islet like or nerve like functions.
  • the present invention provides a method for i nd ucing differentiation of mam mal ian em bryonic stem cells into functioning cells, which comprises the steps of;
  • bFGF basic fibroblast growth factor
  • functioning cells such as pancreatic islet like cell clusters and nerve like cells can be differentiated from the mammalian ES cells.
  • pancreatic islet like cell clusters induced by the present invention have an ability to produce insulin and to secrete insulin in response to glucose stimulation, and the cells consisting the clusters express pancreatic-related endocrine and exocrine markers including insulin, glucagon,
  • Glut-2 islet amyloid polypeptide, amylase and carboxypeptitase.
  • the nerve like cells induced by the present invention exhibit nerve fiber like appearance and the cells consisting the clusters express nerve related markers including nestin, ⁇ -tublin III, seletonin, tyrosin hydroxylase Nurt 1.
  • the inventors grafted the insulin-secreting islet like cell clusters induced from mouse ES cells by the method of the present invention into streptozotocine induced diabetic mice, and succeeded in decreasing the high blood glucose levels of the diabetic mice to those around the normal level. This study supports that the insulin producing islet like cell clusters obtained by the invention are useful for treatment of diabetics.
  • the present invention further provides a method for treating a mammalian patient having disorders in pancreatic islet function, which comprises the step of transplanting islet-like cell clusters induced from allogenic ES cells according to the invention to the patient.
  • the present invention also provides a method for treating a patient with nerve degenerative disease or spinal cord injury, which comprises the step of transplanting nerve like cells induced from allogenic ES cells according to the present invention to the patient.
  • the present invention also provides functioning cells including pancreatic islet like cell clusters and nerve like cells derived from the mammalian ES cells by the method of the present invention.
  • the functioning cells are useful not only for cell transplant therapy but also for in vitro screening of various new drugs which affect or restore islet or nerve function, safety evaluation of new drugs and so on.
  • BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 is a schematic description of differentiation steps of the present invention from ES cells to functioning cells.
  • Fig. 2 represents result of insulin secretion from the cell clusters obtained in Example 1 in response to glucose stimulation.
  • column L represents the amount of insulin secreted per cluster in response to low dose (3.3mg/L) glucose stimulation determined 5 minutes and 30 minutes respectively after the stimulation.
  • Column H represents the amount in response to high dose (25mmol/L) glucose stimulation.
  • Fig. 3 represents time-course of non-fasting blood glucose levels of diabetic mice implanted with the pancreatic islet like cell clusters derived from mouse ES cells compared to that of sham operation group.
  • Fig. 4 represents time course of body weight of diabetic mice implanted with the pancreatic islet like cell clusters derived from ES cells compared to that of sham operation group.
  • Fig. 5 represents percentages of TuJ-positive cell colonies induced from mouse ES cells by culturing in the presence of O(control), 5 and 10mM nicotineamide in steps 3 and 4.
  • Fig. 6 represents percentages of TuJ-positive cell colonies induced from mouse ES cells by culturing in the presence or absence of 4 ng/ml of bFGF (treated or untreated respectively) and then, in the presence of 10 mM nicotineamide (steps 3 and 4).
  • Fig. 7 is a histogram summarizing the effect of bFGF on the proportion of EBs (in step 4) displaying different degrees of network complexity.
  • Fig. 8 represents effects of BMP-4 and FBS on expression of TuJ on the colonies induced from mouse ES cells.
  • Fig. 9-11 represent effects of transplantation of the differentiated and undifferentiated ES cells on 6-OHDA lesioned rats.
  • embryonic stem cell(s) or "ES cell(s)” represents pluripotent cells derived from the inner cell mass of in vitro fertilized blastocytes.
  • Embryoid body or EB represents a cell cluster composed of three embryonic germ layers and formed from ES cells on their in vitro aggregation.
  • the feeder cell layer as used herein is constructed in accordance with procedures known in the art, and may be prepared from mice fatal fibroblast cells. Feeder cells are now, commercially available.
  • the mammalian ES cells which may be used herein are not limited and may be rodent, such as mouse ES cells and rat ES cells, as well as primate such as cynomolgus ES cells and human ES cells. At present, various ES cells are derived and available including mice and human EC cells. Alternatively, the ES cells used herein may be those obtained from mammalian fertilized ovum by means of previous reports. For example, techniques for isolating stable cultures of human embryonic stem cells have been described by Thomson et al. (U.S. patent Nos. 5,843,780 and 6,200,806; Science vol. 282 1145-1147 (1998), the disclosure of these publications are herein incorporated by reference).
  • Step 1 of the present method is a conventional ES cell propagation step, which is described in, such as, N. Lumelsky et al., Science 292, 1389-1394 (2001), the disclosure of the publication is herein incorporated by reference.
  • mouse fetal feeder cells are cultured on a gelatin coated cell culture container to give a layer on the inner surface, then the ES cells are plated on the layer and cultured with an ES cell proliferating medium comprising leukemia inhibiting factor (hereinafter, referred to as "LIF").
  • LIF leukemia inhibiting factor
  • ES cells proliferate in an undifferentiated state.
  • feeder cells may be those commercially available cells or those derived from mice fetal fibroblast cells by a conventional manner.
  • the ES cell proliferating medium used in step 1 may comprise 100-10000U/ml of LIF.
  • a medium used in this step any known medium that contains LIF and is useful for ES cell proliferation can be employed.
  • An especially preferable medium is high glucose Dulbecco's modified
  • LIF leukemia inhibitor factor
  • step 1 culture of the ES cells may be continued until a desired amount of the cells is obtained. Typically, 3-7 days culture may provide enough cells. The obtained
  • ES cells are transferred to the next step.
  • culture of the cells or cell clusters may be carried out under a conventional cell culture condition such as at 37°C, in a humidified atmosphere of 5% CO 2 in 95% air.
  • step 2 the proliferated ES cells are kept in suspension culture with a medium supplemented with LIF and bFGF.
  • LIF has been believed to help retain the ES cells in an undifferentiated state and the art has believed that it is indispensable to exclude LIF from the culture in order to induce differentiation of the ES cells.
  • all of the proposed EB inducing conditions contain the step culturing the expanded ES cells in suspension culture with a medium containing no LIF to allow their aggregation (for example, Su-Chen Zhang et al., Nature biotechnology, 19, 1129-1133 (2001), the disclosure of the publication is herein incorporated by reference).
  • the present inventors succeeded to provide highly efficient EB formation from the ES cells with a medium comprising LIF and bFGF.
  • the medium used in step 2 contains LIF and bFGF.
  • the amount of LIF in the medium may preferably be about 100-10000U/ml.
  • the amount of bFGF in the medium may preferably be about 2-100ng/ml.
  • the medium used in this step may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol and nicotinamide, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, laminin, collagen and heparin in a conventional chemically defined cell culture medium.
  • growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol and nicotinamide, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibro
  • An example of especially preferred medium used in this step is high glucose Dulbecco's modified Eagle's medium (Life Tech.) supplemented with 20% fetal bovine serum replacement (Life Tech.), 2% nonessential amino acid (Life Tech.), 0.1mmol/l 2-mercaptoethanol (Life Tech.), 1000 U/ml of leukemia inhibitor factor (LIF; Life Tech.), 2mmol/l of L- glutamine (Life Tech.) and 4ng/ml of bFGF (R&D systems, Minneapolis).
  • high glucose Dulbecco's modified Eagle's medium (Life Tech.) supplemented with 20% fetal bovine serum replacement (Life Tech.), 2% nonessential amino acid (Life Tech.), 0.1mmol/l 2-mercaptoethanol (Life Tech.), 1000 U/ml of leukemia inhibitor factor (LIF; Life Tech.), 2mmol/l of L- glutamine (Life Tech.) and 4ng/ml of bFGF (R&D systems, Minneapolis).
  • step 2 the ES cells are cultured in suspension without the feeder cell layer to allow the cells aggregate to give embryoid bodies.
  • the formation of EBs may be microscopically monitored. According to the present invention, EB formation may be observed from 2 days of the suspension culture. The suspension culture may be continued for 5-10 days to obtain enough amount of EBs. According to the present invention, a significantly larger number of vital EBs are induced than those induced by a conventional suspension culture step with a medium containing no LIF nor bFGF.
  • step 3 The EBs obtained in step 2 are then transferred to a selection-expansion step (step 3).
  • step 3 thus obtained EBs are plated on a culture container of which inner surface is coated with a protein, such as collagen type IV, and cultured with an appropriate selection-expansion medium. It is preferable to culture the EBs in the protein coated container with the medium used in step 2 for about 2 days and then exchange the medium with a selection- expanding medium.
  • the selection-expanding medium used in step 3 may preferably be a serum-free cell culture medium supplemented with nicotinamide, insulin and fibronectine.
  • the medium used in this step may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as laminin, collagen and heparin in a conventional chemically defined cell culture medium.
  • growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as laminin, collagen and heparin in a conventional chemically defined cell culture medium.
  • preferable medium is a serum free DMEM/F-12 medium supplemented with nicotinamide, fibronectine, and N-2 supplements (GIBCO, 17502-014: consisting of Insulin 500 ⁇ g/ml, Human transferin 10000 ⁇ g/ml, Progesterone 0.63 ⁇ g/ml, Putrescine 1611 ⁇ g/ml and Selenite 0.52 ⁇ g/ml in water).
  • GEBCO serum free DMEM/F-12 medium supplemented with nicotinamide, fibronectine, and N-2 supplements
  • the EBs may be cultured with the selection- expanding medium for 3-14 days, preferably for 4-7 days.
  • the cell clusters obtained in step 3 are then dissociated from the container and plated on a culture container of which inner surface is coated with a protein or an amino acid.
  • the transferred clusters are further cultured in a differentiation medium.
  • step 4 the cell clusters can be differentiated into either pancreatic islet like cell clusters or nerve like cells.
  • the cell clusters may be cultured with a serum-free cell culture medium supplemented with nicotinamide, insulin and laminine.
  • the medium may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, collagen and heparin in a conventional chemically defined cell culture medium.
  • growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, collagen and heparin in a conventional chemically defined cell culture medium.
  • An especially preferred example is serum-free DMEM/F12 medium supplemented with nicotinamide, lamin
  • the cell clusters may be cultured with a serum-free cell culture medium supplemented with lysine and laminine.
  • the medium may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol and nicotinamide, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, collagen and heparin in a conventional chemically defined cell culture medium.
  • growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol and nicotinamide, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, collagen and heparin in a conventional chemically defined cell culture medium.
  • An especially preferred example is serum-free DMEM/F12 medium supplemented
  • cell clusters may be cultured for 3-90 days or longer. 4-12 days culture will be enough for differentiation into the desired functioning cells and further culture may provide further proliferation of the differentiated clusters.
  • pancreatic islet like cell clusters obtained by the present invention represent an ability to produce insulin and to secret insulin in response to glucose stimulation, and the cells consisting the clusters express genes specific to pancreatic endocrine cells including insulin, glucagon, Glut-2 and islet amyloid polypeptide as well as those specific to pancreatic exocrine cells including amylase and carboxypeptitase.
  • the nerve like cells obtained by the present invention represent nerve fiber like appearances and express markers relevant to nerve cells including nestin, b-tublin III, seletonin, tyrosine hydroxylase. Therefore, said nerve like cells are capable of generating mature neurons.
  • mice ES cells as well as human ES cells proliferate in vitro in an undifferentiated state retaining the pluripotency for more than one year, the present method can be employed to provide enough amount of donor cells used in the cell transplanting therapy.
  • the present invention further provide a method for treating a mammalian patient having disorders in pancreatic islet function, which comprises transplanting islet-like cell clusters induced from allogenic ES cells according to the invention to the patient.
  • a mammalian patient having disorders in pancreatic islet function which comprises transplanting islet-like cell clusters induced from allogenic ES cells according to the invention to the patient.
  • pancreatic islet function includes, but not limited to, type I diabetic patient, pancreatomized patient and insulin-required diabetic patient such as type II diabetic patient or patient with cystic fibrosis.
  • the mammalian patient may include human patient.
  • transplantation of the pancreatic islet like cell clusters obtained as above may be carried out according to a clinically performed or proposed islet transplantation protocol (for example, Kazutomo Inoue and Masaaki Miyamoto, J. Hepatobiliary Pancreat. Surg.
  • the pancreatic islet like cell clusters may be implanted intraportally into the liver.
  • the pancreatic islet like cell clusters may be implanted into a prevascularized subcutaneous site.
  • Said clusters may be macroencapsulated with a bio-compatible material before implantation. The amount of the clusters to be transplanted will be determined by the art based on the titer of the obtained clusters as well as the general conditions, age, sex, body weight of the patient to be treated.
  • the present invention also provides a method for treating a mammalian patient having disorders in nerve function, which comprises a step of transplanting the nerve like cells derived from allogenic ES cells to the patient.
  • mammalian patient having disorders in nerve function includes, but not limited to, patients having nerve degeneration disease such as Alzheimer's disease and Creutzfeldt-Jakob disease or spinal injury.
  • the mammalian patient may include human patient.
  • Example 1 The present invention will be further illustrated by the following Examples. The examples are intended to illustrate but not in any means to limit the invention. Example 1
  • pancreatic islet like clusters from mouse
  • mouse ES cell line 129sv (passages 11; Dainippon Pharmaceutical Co. Ltd., Osaka Japan) was used.
  • mouse ES cell clonal line derived from C57/BL6 mouse (passage 11; kindly provided by Professor Norio NAKATSUJI of Institute for frontier medical sciences, Kyoto University, Kyoto Japan) and similar results as below were obtained (data not shown).
  • Mammalian ES cells can be proliferated in an undifferentiated state if they are cultured on a feeder layer in the presence of leukemia inhibitor factor.
  • Mouse embryo feeder cells (Dainippon Pharmaceutical Co.
  • ES cell culture medium of high glucose Dulbecco's modified Eagle's medium (D-MEM Cat# 12100: Life Technology, Grand, NY) supplemented with 20% fetal bovine serum replacement (Life Tech.), 2% nonessential amino acid (Life Tech.), 0.1mmol/l 2- mercaptoethanol (Life Tech.), 1000 U/ml of leukemia inhibitor factor (LIF; Life Tech.) and 2mmol/l L- glutamine (Life Tech.) was used.
  • a feeder layer of the mitomycin treated mouse embryonic fibroblasts was prepared on a gelatin-coated culture dish (6 cm), 5 ml of the medium was added thereto and 10 6 of ES cells were plated on the layer. Cells were cultured at 37°C in humidified atmosphere of 5% CO 2 in 95% air. Every 3 days, the cells were removed from the dish by means of 0.05% trypsin solution in 0.04% EDTA (Life Tech.) and passaged into a freshly prepared medium on a freshly prepared feeder layer. The ES cells were cultured for 3-7 days. Step 2
  • EBs embryoid bodies
  • the ES cells were disassociated by means of the trypsin-EDTA solution and were plated on a non-adherent culture dish to give cell density of 6 x 10 5 cells/cm 2 .
  • the cells were kept in suspension culture in the medium used in the above step 1 in the absence or presence of bFGF (4 ng/ml; R&D Systems, Minneapolis, U.S.A. and Kaken Pharmaceuticals, Co. Ltd., Tokyo Japan). Cells were cultured at 37°C in humidified atmosphere of 5% CO 2 in 95% air. Every 2 days, the media were replaced with freshly prepared ones.
  • the EBs obtained in step 2 with the bFGF containing medium were plated on a Type IV collagen (Sigma, St. Louis, MO) coated 6cm dish filled with the medium used in step 2.
  • the medium was replaced with selection-expanding medium of serum free DMEM/F- 12(1:1) medium (cat# 11320, Life Tech.) supplemented with 500 ⁇ g/ ml of Bovine Insulin, 1 ⁇ g / ml of Progestron, 1600 ⁇ g /ml of Putrescine and 5 ⁇ g / ml of Fibronectin and 10mM of nicotinamide.
  • the cell clusters were cultured for more than 7 days.
  • Step 4 Differentiation of the cells After 7 days culture with the selection- expanding medium, further differentiation was induced by culturing the cell clusters with serum free DMEM/F-12(1 :1 ) medium (cat# 11320, Life Tech.) supplenmented with 500 ⁇ g/ ml of Bovine Insulin, 1 ⁇ g/ ml of Progestron, 1600 ⁇ g /ml of Putrescine, 1 ⁇ g / ml of Laminin and 10mM of nicotinamide. The cells were incubated at 37°C in humidified atmosphere of 5% CO 2 in 95% air for 12 days to give islet like cell clusters of about 100-400 ⁇ m in diameter. RNA extraction and RT- PCR analysis At the end of every step as above, pancreatic relating gene expression on the cells was examined by means of RT-PCR analysis.
  • RNA of the cells obtained in each step was. isolated using ISOGEN (Nippon Gene; Osaka, Japan) according to the manufacturer's instruction. The cells were homogenized in 0.8 ml of ISOGEN using a Potter homogenizer at 4°C. The homogenate was mixed with 1 ml of chloroform, and RNA in the aqueous phase was precipitated with the same volume of isopropyl alcohol. Synthesize of cDNA was carried out with oligo dT primers (Takara Shuzo Co. Ltd., Kyoto, Japan) and Moloney murine leukemia virus (M-MLV) Superscript II reverse transcriptase (Gibco/BRL) following the manufacturer's instructions.
  • ISOGEN Natural Gene
  • M-MLV Moloney murine leukemia virus
  • telomere length a parameter indicative of transcription factor mRNAs.
  • PCR was carried out using standard protocols with Taq polymerase (Boehringer-Mannheim, Indianapolis, IN). Cycling parameters were as follows, denaturation at 94°C for 1min, annealing at 52-61°C for 30- 120 seconds (depending on the primer) for 1min, and elongation at 72°C for 1 min. The number of cycles varied between 25 and 40, depending on the particular mRNA abundance. The number of cycles and the amount of cDNA were chosen in such a way as to select PCR conditions on the linear portion of the reaction curve avoiding "saturation effects" of PCR. Obtained PCR products were confirmed by sequencing.
  • Primer sequences forward an reverse
  • length of the amplified products were as follows: ⁇ -actin:
  • IAPP Islet Amyloid Polypeptide
  • Glucose transporter 2 (Glut2): CCACCCAGTTTACAAGCTC
  • HNF3 ACCTGAGTCCGAGTCTGACC GGCACCTTGAGAAAGCAGTC
  • pancreatic transcription factor PDX-1 which is indispensable for pancreatic development, was expressed in steps 1 and 4 cells.
  • Oct-4 which relates to differentiation of ES cell, was expressed in step 1 cells and down- regulated with the differentiation of the ES cells.
  • the ES cells at every step expressed a maker of definitive (embryonic) and visceral (extra-embryonic) endoderm GATA-4 and definitive endoderm HNF3 ⁇ concerning markers of pancreatic ⁇ cell fate.
  • Nestin a transcription factor relates to immature hormone-negative pancreatic cells, was strongly expressed in step 2 and down-regulated with the differentiation of the ES cells. The results showed that many nestin positive progenitor cells were contained in the EBs obtained in the presence of bFGF.
  • EBs induced by bFGF treatment expressed transcription factors of endocrine (Insulin I, Insulin II, Glucagon, Glucose transporter-2 (Glut-2) and Islet Amyloid Polypeptide) specific genes whereas any gene concerning pancreatic islet cells did not expressed in the cell clusters obtained in step 2 using the medium without bFGF.
  • the cells expressed exocrine specific genes (amylase and carboxypeptidase).
  • pancreatic islet like cell clusters of the invention can be matured to a pancreatic tissue structure, which composed of endocrine cells including glucagon-producing ⁇ cells, insulin- producing ⁇ cells, pancreatic polypeptide-producing y cells, and somatostin-producing ⁇ cells and exocrine cells.
  • the cell clusters obtained in step 4 (20-25 clusters) were washed 3 times with PBS(-) and plated on a 6cm cell culture dish containing Krebs-Ringer with bicarbonate buffer consisting of 120 mM NaCl, 5 mM KCI, 2.5 mM CaCI 2 , 1.1 mM MgCI 2 , 25 mM NaHC0 3 and 0.1% bovine serum albumin, and incubated at 37°C. 3.3 mmol/l (L) or 25mmoi/l (H) glucose was added thereto and incubated. Five and thirty minutes after the glucose stimulation, the insulin contents in the buffer were measured using insulin enzyme- linked immunosorbence assay (ELISA) kit (ALPCO, Windham, NH). Results are shown in Figure 2. In the Fig. 2, the amounts of insulin secreted per one cluster in response to the low or high glucose stimulation at 5 and 30 minutes after the stimulation are shown. The clusters exhibited insulin secretion in response to glucose stimulation in a dose dependent manner.
  • ELISA insulin enzyme
  • the cell clusters obtained in step 4 were extracted with acid ethanol (10% glacial acetic acid in absolute ethanol) overnight at 4°C, followed by cell sonication and then, the insulin content in the supernatant was determined by means of the ELIZA kit.
  • Total cellular protein amount was determined using DC protein assay system (Bio-Rad laboratories, Hercules, CA). The total cellular insulin content of those cell clusters was 71.3ng/mg protein.
  • Paraffin slices of the cell clusters obtained in step 4 were prepared as follows.
  • the cell clusters (in step 4, incubated 12 days) were washed three times with ice-cold PBS and were fixed with methanol/aceton (1:1) for over night.
  • the clusters were dehydrated with aqueous alcohol (70-100%), then embedded in a paraffin block and the block was sliced to give 4 ⁇ m and 8 ⁇ m thick slices.
  • fluorescently labeled secondary antibodies Jackson Immunoresearch Laboratories, West Grove, PA were used according to the supplier's instruction.
  • the obtained insulin producing cell cluster was strongly positive to insulin and glucagone, and positive to nestin and TuJ1.
  • Example 2 Transplantation of the insulin producing cell clusters into STZ derived diabetic mice.
  • pancreatic islet like cell clusters were transplanted to determine if the cluster could differentiate into functioning pancreatic islet in vivo. All animal studies were carried out in accordance with
  • mice were prepared according to the method disclosed in H. Iwata et al., Pancreas vol. 23(4) 375-381(2001), the disclosure of the publication is herein incorporated by reference.
  • Streptozotocin (STZ) cryopreserved powder (Sigma, St. Louis, MO) was dissolved in 0.1 M citrate buffer, pH 4.5 before use.
  • the STZ solution was intraperitoneally injected (227 mg/kg of body weight) to 8- to 10-weeks-old male Nude mice (Shimizu, Kyoto, Japan), Stable hyperglycemia, i.e. increased blood glucose levels of about 350 - 600 mg/dl) were usually developed 7 to 10 days after the STZ single injection.
  • Blood glucose level of the mouse was determined using Glucometer Elite .
  • XL blood glucose meter (Fujii Corp., Tokyo, Japan). Animals represent 350mg/dl or more non- fasting blood glucose at 7-10 days of STZ injection were regarded as diabetic mice and used at 14 day from the STZ injection.
  • the diabetic animals were grafted with 3000 insulin producing pancreatic islet like cell clusters obtained in Example 1 or received sham operation.
  • the cell clusters suspended in PBS(-) were injected into the kidney subcapsuler region (one kidney) of the diabetic mice with 23-gauge winged needle.
  • the same volume of PBS(-) was injected in the same manner as above.
  • the experimental group received cell clusters, non-treated control group and sham group consisted of 6, 3 and 3 animals respectively. After the transplantation, non-fasting blood glucose and body weight were monitored daily. The results are shown in Figures 3 and 4.
  • the blood glucose of the experimental group significantly decreased and the significantly lower blood glucose level than the sham group was kept throughout the time of the experiment.
  • the body weight of the implanted group increased slightly and kept stable.
  • mice implanted with the clusters were sacrificed respectively. All implanted mice remained healthy until killed and kept significantly lower blood glucose than the sham group. To the contrarily, blood glucose levels in non-treated control and sham groups were increased gradually and became exhausted. All of the mice of control and sham groups died prematurely from complication of diabetics between day 14 and day 30 of the operation.
  • tissue slices of 4-8 ⁇ m thickness were immunohistochemically examined in the same manner as Example 1.
  • the mouse ES cells same as used in Example 1 were treated in the same manner as steps 1-3 of Example 1.
  • Thus obtained cell clusters were then cultured in a dish coated with poly-L-lysine and filled with serum free DMEM/ F-12(1:1) medium (cat# 11320, Life Tech.) supplemented with 500 ⁇ g/ ml of Bovine Insulin, 1 ⁇ g/ ml of Progestron, 1600 ⁇ g /ml of Putrescine, 10mM of lysine and 1 ⁇ g / ml of Laminin.
  • the cells were cultured for 12 days and the obtained cells were examined genetically and immunohistochemically according to the same manner as described in Example 1.
  • thyrosine hydroxylase(TH) polyclonal 1:200 (Pel-Freeze, Rogers, AR), thyrosine hydroxylase(TH) monoclonal 1:1000 (Sigma, St. Louis, MO), serotonin polyclonal 1:4000 (Sigma, St. Louis, MO), MAP 2 polyclonal (Chemicon International, Temecula, CA), and GFAP monoclonal (Clon Tech, Palo Alto, CA) were used in addition to the antibodies used in Example 1.
  • the obtained cells represented nerve fiber like appearance and were immunohistochemically positive to nestin, TuJ1 ( ⁇ -tublin III), serotonin, GFAP, MAP 2 and tyrosine-hydroxylase.
  • TuJ1 ⁇ -tublin III
  • serotonin GFAP
  • MAP 2 MAP 2
  • tyrosine-hydroxylase a cell proliferation factor.
  • the primer sequences used herein for detecting Nurt-1 were as follows: TGAAGAGAGC GGAGAAGGAG ATC TCTGGAGTTA AGAAATCGGA GCTG 255 bp.
  • the obtained nerve like cells are capable of generating mature neurons if they are implanted in vivo.
  • insulin producing pancreatic islet like cell clusters are obtained from human ES cells.
  • ES cells may be those described in the art such as U.S. patent Nos. 5,843,780 and 6,200,806; Science 282, 1145-1147 (1998), the disclosures of the publications are herein incorporated by reference.
  • the cell clusters produce insulin and secret insulin in response to glucose in a dose dependent manner.
  • the cell clusters are implanted into a human type I diabetic patient. About 3-5 10 5 clusters are suspended in about 50-100 ml of Krebs-Ringer solution and the suspension is injected into the liver via portal vein, or is implanted to subcutaneous space as a bio-artificial pancreas. The pancreatic function of the implanted patient restores and the patient acquires insulin-independency.
  • the cells were propagated on a feeder layer of mitomycin treated mouse embryonic fibroblasts in DMEM (Invitrogen, Grand Island, NY, USA) medium containing 20% fetal bovine serum (FBS, JRH Bioscience, Lenexa, KS, USA), 1000 unit/ml leukimia inhibitory factor (LIF), 50mM 2- mercaptoethanol, 2mM L-glutamic acid and 10X non- essential amino acid (Invitrogen, Grand Island, NY, USA).
  • ES cell colonies were detached and subcultured until a desired amount of the cells is obtained.
  • EB culture medium used in the above step.
  • the EB culture medium was replaced with serum free DMEM/F-12 (Invitrogen, Grand Island, NY ⁇ USA) medium containing 500 ⁇ g/ml of bovine insulin, 1 ⁇ g/ml of progestron, 1600 ⁇ g/ml of putrescine, 0, 5 or 10mM of nicotinamide and 5 ⁇ gl ⁇ of fibronectin (Nakalai tesque, Kyoto), and cultured for 4 days.
  • serum free DMEM/F-12 Invitrogen, Grand Island, NY ⁇ USA
  • Step 4 After 4 days of culture, cell differentiation were induced by replacing the medium with serum free DMEM/F- 12 medium containing 500 ⁇ gl ml of bovine insulin, 1 ⁇ g/ml of progestron, 1600 ⁇ g/ml of putrescine, 0, 5 or 10mM of nicotinamide, 5 ⁇ gl ml of fibronectin (Nakalai tesque, Kyoto, Japan) and 1 ⁇ gl ml of laminin (Nakalai tesque, Kyoto, Japan). The cells were cultured and obtained cells were examined as follows.
  • the undifferentiated ES cells (step 1) and those obtained in step 4 were genetically examined.
  • the examined cells were those treated with bFGF in step 2 and cultured in the presence of 10mM NA in steps 3 and 4.
  • the primers used in this examination are follows:
  • Nurt-1 tyrosin hydroxylase TGAAGAGAGC GGAGAAGGAG ATC TCTGGAGTTA AGAAATCGGA GCTG
  • Wnt-1 Mesencephalic domaminergic neuron transcription factor signaling molecule
  • Engrailed-1 (En 1): Mesencephalic domaminergic neuron transcription factor
  • Otx 1 Transcription factor critical in brain morphogenesis
  • Otx 2 Transcription factor critical in brain morphogenesis ⁇ '-CCATGACCTATACTCAGGCTTCAGG-S'
  • Fibroblast growth factor receptor (FGFR 3R)
  • Fig. 7 is histogram summarizing the effect of NA (nicotineamide) and bFGF on complexities of the neural network like appearance of the EBs.
  • the EBs were cultured in the presence or absence of 4 ng/ml of bFGF in step 2 (indicated "non-treated” or “treated” respectively) and then cultured in the presence of NA in steps 3 and 4. Each condition was assayed by counting all EBs in culture and the experiment was repeated twice resulting in similar results.
  • BMP-4 or FBS treatment The cells treated with bFGF in step 2 were then cultured in the presence of 0.5 nM bone morphogenetic protein (BMP-4) or 5% fetal bovine serum (FBS) in addition to 10mM of NA in the following steps (steps 3 and 4). In this procedure, BMP4 was freshly added at each medium change. Both BMP-4 and FBS suppress differentiation of the cells into neural cells.
  • BMP-4 bone morphogenetic protein
  • FBS fetal bovine serum
  • 6-OHDA Parkinson's disease
  • PD Parkinson's disease
  • marked dopamine depletion (over 95%) in the striatum induces the supersensitivity in dopamine receptors.
  • 6-OHDA-lesioned rats therefore, a mixed D1/D2 receptor agonist apomorphne induces rotational asymmetry in the contralateral direction, while a DA releaser amphetamine induces the ipsilateral rotation. Accordingly, we studied in vivo effects of transplantation of the differentiated and undifferentiated ES cells on the 6-OHDA lesion model.
  • the rats were injected with the 6-OHDA solution into left substantia nigra, where the bregma was -4.8 mm caudal, 1.8 mm left lateral, -7.8 mm ventral, via a motor-driven 10 ⁇ l-Hamilton syringe using 26 gauge needle. Coordinates were set according to the rat brain atlas (Paxinos G. and Watson C. (1986) "The rat brain in stereotaxic coordinates" (Second Edition). Academic Press, North Ryde, Australia). After 2 weeks, lesioned rats were selected for transplantation by estimation of rotational behavior in response to apomorphine (Sigma).
  • the selected rats were anesthetized (sodium pentobarbital, 50 mg/kg, i.p.) and then placed in Kopf stereotaxic frame. Each animal received a microinjection of 1.0 ⁇ l of grafting cell suspension or the vehicle into two sites of the left striatum, where the bregma was +1.0 mm caudal, 3.0 mm left later, -5.5 and 5.0 mm ventral according to the rat brain atlas (id), using a 10 ⁇ l-Hamilton syringe with 22 gauge needle. A 5-min waiting period allowed transplanted cells to settle before the needle was removed. Subsequently, apomorphine-induced rotational asymmetry was assesses every 2 weeks.
  • Results are shown in Figs. 9-11. Data are presented as mean ⁇ SEM and compared using the unpaired Student's test or one-way analysis of variance. Differences were considered significant at P ⁇ 0.05. Further statistical analysis for post hoc comparison was performed using the Bonferroni/Dunn test (Statview, Abacus Concepts, Berkely, USA) for the numbers of apomorphine-induced rotational asymmetry.
  • nerve like cell can be obtained from human ES cells.
  • Human ES cells may be those described in Example 4. Although some minor modifications would be required for optimizing the culture conditions, it would be mere routine work for the art.
  • dopaminergic neurons can effectively enriched from the total population of differentiated ES cells without gene modification, flow cytometry sorting or separation with magnetic beads.
  • the nerve like cells induced from ES cells by the simple 4-step method of the instant invention represent good therapeutic effect on Perkinson's disease and the patients received the treatment can survive long- term .

Abstract

The present invention provides a 4-step method for inducing differentiation of embryonic stem cells into functioning cells comprising 1) expanding ES cells; 2) inducing Embryoid Bodies in the presence of leukemia Inhibitor factor and basic FGF; 3) selection expanding of the EBs and 4) differentiation. According to the present invention, ES cells can be differentiated into either insulin producing pancreatic islet like cell clusters or nerve like cells. Thus obtained functioning cells may be potential sources of donor cells in cell transplant therapy for many patients.

Description

DESCRIPTION
METHOD FOR INDUCING DIFFERENTIATION OF
EMBRYONIC STEM CELLS INTO FUNCTIONING CELLS
FIELD OF THE INVENTION The present invention relates to a method for inducing differentiation of mammalian embryonic stem cells into functioning cells. The present invention also relates to the functioning cells obtained by the present invention and a method for treatment of a patient by implanting functioning cells to the patient. ART RELATED
Pluripotent stem cells have been derived from two embryonic sources. Embryonic stem (ES) cells are derived from the inner cell mass of preimplantation embryos, and embryonic germ (EG) cells are derived from primordial germ cells (PGCs). Both ES and EG cells are pluripotent and demonstrate germ-line transmission in experimentally produced chimeras. Mouse ES and EG cells share several morphological characteristics such as high levels of intracellular alkaline phosphatase (AP), and presentation of specific cell surface glycolipids and glycoproteins. These properties are characteristic of, but not specific for, pluripotent stem cells. Other important characteristics include growth as multicellular colonies, normal and stable karyotypes, the ability to be continuously passaged, and the capability to differentiate into cells derived from all three embryonic germ layers. Pluripotent stem cell lines that share most of these characteristics also have been reported for chicken, mink, hamster, pig, rhesus monkey, and common marmoset. Also a stem cell is a cell that has the ability to divide (self-replication) for indefinite periods- often throughout the life of the organism. Under the right conditions, or given the right signals, stem cells can give rise (differentiate) to the many different cell types that make up the organism.
Recently, S.H. Lee et al. (Nature Biotechnology 18, 675 - 679 (2000), the disclosure of the publication is herein incorporated by reference) disclosed to generate CNS progenitor populations from ES cells, to expand these cells and to promote their differentiation into dopaminergic and serotonergic neurons in the presence of mitogens and specific signaling molecules. The differentiation and maturation of neuronal cells was completed after mitogen withdrawal from the growth medium. This experimental system provides a powerful tool for analyzing the molecular mechanisms controlling the functions of these neurons in vitro and in vivo, and potentially for understanding and treating neurodegenerative and psychiatric diseases.
Also, H. Kawasaki et al. (Neuron 28, 31-40(2000), the disclosure of the publication is herein incorporated by reference) have identified a stromal cell-derived inducing activity (SDIA) that promotes neural differentiation of mouse ES cells. SDIA accumulates on the surface of PA6 stromal cells and induces efficient neuronal differentiation of co-cultured ES cells in serum-free conditions without use of either retinoic acid or embryonic bodies. BMP4, which acts as an antineuralizing morphogen in Xenopus, suppresses SDIA-induced neuralization and promotes epidermal differentiation. A high proportion of tyrosine hydroxylase-positive neurons producing dopamine are obtained from SDIA-treated ES cells. When transplanted, SDIA-induced dopaminergic neurons integrate into the mouse striatum and remain positive for tyrosine hydroxylase expression. Neural induction by SDIA provides a new powerful tool for both basic neuroscience research and therapeutic applications.
In a study of B. Soria et al., mouse embryonic stem cells have been introduced as a new potential source for cell therapy in type 1 diabetic patients (Diabetes 49: 157- 162 (2000), the disclosure of the publication is herein incorporated by reference). Using a cell-trapping system, they have obtained an insulin-secreting cell clone from undifferentiated ES cells. The construction used allows the expression of a neomycin selection system under the control of the regulatory regions of the human insulin gene. The chimeric gene also contained a hygromycin resistance gene (pGK-hygro) to select transfected cells. A resulting clone (IB/3x-99) containing 16.5ng/μg protein of total insulin displays regulated hormone secretion in vitro in the presence of various secretagogues. Clusters obtained from this clone were implanted in the spleen of streptozotocin- induced diabetic animals. Hyperglycemia of the transplanted animals were normalized within one week and their body weight were restored in 4 weeks. Whereas slower recovery was observed in the transplanted animals than control mice in an intraperitoneal glucose tolerance test, blood glucose levers after meal load were normalized in a similar manner. This approach opens new possibilities for tissue transplantation in the treatment of typel and type 2 diabetes and offers an alternative to gene therapy.
S. Assady et al. (Diabetes 50: 1691-1697 (2001), the disclosure of the publication is herein incorporated by reference), used pluripotent undifferentiated human embryonic stem cells (hES) as a model system for lineage- specific differentiation. They cultured hES cells in both adherent and suspension culture conditions, and observed spontaneous in vitro differentiation of the cells including generation of cells with characteristics of insulin-producing β-cells. Immunohistochemical staining for insulin was observed in a surprisingly high percentage of the cells. Secretion of insulin into the medium was observed in a differentiation-dependent manner and was associated with the appearance of other β-cell markers. These findings suggest that the hES cell model system is a potential basis for enrichment of human β-cells or their precursors, as a possible future source for cell replacement therapy in diabetes.
Su-Chun Zhang et al. (Nature Biotech. 19, 1129-1133 (2001), the disclosure of the publication is herein incorporated by reference) disclose in vitro differentiation, enrichment, and transplantation of neural precursor cells from human ES cells. Upon aggregation to embryoid bodies, differentiating ES cells formed large numbers of neural tube-like structures in the presence of fibroblast growth factor 2 (FGF-2). Neural precursors within these formations were isolated by selective enzymatic digestion and further purified on the basis of differential adhesion. Following withdrawal of FGF-2, they differentiated into neurons, astrocytes, and oligodendrocytes. After transplantation into the neonatal mouse brain, human ES cell-derived neural precursors were incorporated into a variety of brain regions, where they differentiated into both neurons and astrocytes. No teratoma formation was observed in the transplant recipients. These results depict human ES cells as a source of transplantable neural precursors for possible nervous system repair.
Nadya Lumelsky et al. disclose a series of experiments in which they induced mouse embryonic cells to differentiate into insulin-secreting structures that resembled pancreatic islet (Science 292, 1389-1394 (2001), the disclosure of the publication is incorporated herein by reference). They have generated cells expressing insulin and other pancreatic endocrine hormones from mouse ES cells. The cells self-assemble to form three-dimensional cluster similar in topology to normal pancreatic islets where pancreatic cell types are in close association with neurons. Glucose triggers insulin release from these cell clusters by mechanisms similar to those employed in vivo. When injected into diabetic mice, the insulin-producing cells undergo rapid vascularization and maintain a clustered, islet-like organization.
However, the insulin-producing cells obtained by Lumelsky did not express pancreatic specific markers, amylase and carboxypeptidase. Further, Lumelsky grafted the insulin-producing cells into a diabetic model animal but failed to observe a sustained correction of hyperglycemia in the model animal.
Seven million people in Japan and 16 million people in the United States are affected by type I diabetics. At present, daily insulin administration or allogenic pancreas transplantation is em ployed for treatment of diabetics. Although the overal l success rates of the pancreas transplantation have significantly increased , organ transplantation requires very invasive surgery and life-long immunosurpressive treatments, wh ich significantly strain the patient. Further, availability of donor organs is stil l serious problem preventing the operation to be popular. Therefore, development of a simple and universal treatment for diabetes is desired. SU MMARY OF TH E I NVENTION
One object of the present invention is to provide a novel method for inducing differentiation of pluripotent embryonic stem cel ls into functioning cells, especially pancreatic islet like cell clusters and nerve l ike cells. Another object of the present invention to provide a method for treating a patient having disorders in pancreatic islet function.
Another object of the present invention is to provide a method for treating a patient having neuronal degeneration or spinal code disorders.
Further object of the present i nvention is to provide functioning cells whi ch are derived from mammalian ES cel ls and exhibit pancreatic islet like or nerve like functions.
Accordingly, the present invention provides a method for i nd ucing differentiation of mam mal ian em bryonic stem cells into functioning cells, which comprises the steps of;
1) culturing the mammalian embryonic stem cells together with feeder cells with a medium comprising leukemia inhibitor factor; 2) culturing the obtained cells in absence of feeder cells with a medium comprising leukemia Inhibitor factor and basic fibroblast growth factor (hereinafter referred to as "bFGF") in suspension culture condition to give embryonic bodies; 3) culturing the obtained embryonic bodies with a growth-selection medium; and
4) culturing the obtained cell clusters with a differentiation medium to give functioning cells.
According to the present invention, functioning cells such as pancreatic islet like cell clusters and nerve like cells can be differentiated from the mammalian ES cells.
The pancreatic islet like cell clusters induced by the present invention have an ability to produce insulin and to secrete insulin in response to glucose stimulation, and the cells consisting the clusters express pancreatic-related endocrine and exocrine markers including insulin, glucagon,
Glut-2, islet amyloid polypeptide, amylase and carboxypeptitase.
The nerve like cells induced by the present invention exhibit nerve fiber like appearance and the cells consisting the clusters express nerve related markers including nestin, β-tublin III, seletonin, tyrosin hydroxylase Nurt 1.
The inventors grafted the insulin-secreting islet like cell clusters induced from mouse ES cells by the method of the present invention into streptozotocine induced diabetic mice, and succeeded in decreasing the high blood glucose levels of the diabetic mice to those around the normal level. This study supports that the insulin producing islet like cell clusters obtained by the invention are useful for treatment of diabetics.
The present invention further provides a method for treating a mammalian patient having disorders in pancreatic islet function, which comprises the step of transplanting islet-like cell clusters induced from allogenic ES cells according to the invention to the patient.
The present invention also provides a method for treating a patient with nerve degenerative disease or spinal cord injury, which comprises the step of transplanting nerve like cells induced from allogenic ES cells according to the present invention to the patient.
Further, the present invention also provides functioning cells including pancreatic islet like cell clusters and nerve like cells derived from the mammalian ES cells by the method of the present invention. The functioning cells are useful not only for cell transplant therapy but also for in vitro screening of various new drugs which affect or restore islet or nerve function, safety evaluation of new drugs and so on. BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 is a schematic description of differentiation steps of the present invention from ES cells to functioning cells.
Fig. 2 represents result of insulin secretion from the cell clusters obtained in Example 1 in response to glucose stimulation. In this graph, column L represents the amount of insulin secreted per cluster in response to low dose (3.3mg/L) glucose stimulation determined 5 minutes and 30 minutes respectively after the stimulation. Column H represents the amount in response to high dose (25mmol/L) glucose stimulation.
Fig. 3 represents time-course of non-fasting blood glucose levels of diabetic mice implanted with the pancreatic islet like cell clusters derived from mouse ES cells compared to that of sham operation group. Fig. 4 represents time course of body weight of diabetic mice implanted with the pancreatic islet like cell clusters derived from ES cells compared to that of sham operation group.
Fig. 5 represents percentages of TuJ-positive cell colonies induced from mouse ES cells by culturing in the presence of O(control), 5 and 10mM nicotineamide in steps 3 and 4.
Fig. 6 represents percentages of TuJ-positive cell colonies induced from mouse ES cells by culturing in the presence or absence of 4 ng/ml of bFGF (treated or untreated respectively) and then, in the presence of 10 mM nicotineamide (steps 3 and 4).
Fig. 7 is a histogram summarizing the effect of bFGF on the proportion of EBs (in step 4) displaying different degrees of network complexity.
Fig. 8 represents effects of BMP-4 and FBS on expression of TuJ on the colonies induced from mouse ES cells.
Fig. 9-11 represent effects of transplantation of the differentiated and undifferentiated ES cells on 6-OHDA lesioned rats. DETAILED DESCRIPTION OF THE INVENTION
In the specification, claims and drawings of the instant application, the term "embryonic stem cell(s)" or "ES cell(s)" represents pluripotent cells derived from the inner cell mass of in vitro fertilized blastocytes.
Embryoid body or EB represents a cell cluster composed of three embryonic germ layers and formed from ES cells on their in vitro aggregation. The feeder cell layer as used herein is constructed in accordance with procedures known in the art, and may be prepared from mice fatal fibroblast cells. Feeder cells are now, commercially available.
The mammalian ES cells which may be used herein are not limited and may be rodent, such as mouse ES cells and rat ES cells, as well as primate such as cynomolgus ES cells and human ES cells. At present, various ES cells are derived and available including mice and human EC cells. Alternatively, the ES cells used herein may be those obtained from mammalian fertilized ovum by means of previous reports. For example, techniques for isolating stable cultures of human embryonic stem cells have been described by Thomson et al. (U.S. patent Nos. 5,843,780 and 6,200,806; Science vol. 282 1145-1147 (1998), the disclosure of these publications are herein incorporated by reference).
Step 1 of the present method is a conventional ES cell propagation step, which is described in, such as, N. Lumelsky et al., Science 292, 1389-1394 (2001), the disclosure of the publication is herein incorporated by reference.
Typically, mouse fetal feeder cells are cultured on a gelatin coated cell culture container to give a layer on the inner surface, then the ES cells are plated on the layer and cultured with an ES cell proliferating medium comprising leukemia inhibiting factor (hereinafter, referred to as "LIF").
By culturing under such condition as above, ES cells proliferate in an undifferentiated state.
In the method of the present invention, feeder cells may be those commercially available cells or those derived from mice fetal fibroblast cells by a conventional manner.
The ES cell proliferating medium used in step 1 may comprise 100-10000U/ml of LIF. As a medium used in this step, any known medium that contains LIF and is useful for ES cell proliferation can be employed. An especially preferable medium is high glucose Dulbecco's modified
Eagle's medium (Life Technology (herein below, Life Tech,),
Grand, NY) supplemented with 20% fetal bovine serum replacement (Life Tech.), 2% nonessential amino acid (Life Tech.), 0.1mmol/l 2-mercaptoethanol (Life Tech.), 1000
U/ml of leukemia inhibitor factor (LIF; Life Tech.) and
2mmol/l L- glutamine (Life Tech.).
In step 1, culture of the ES cells may be continued until a desired amount of the cells is obtained. Typically, 3-7 days culture may provide enough cells. The obtained
ES cells are transferred to the next step.
Throughout the inducing method of the present invention, culture of the cells or cell clusters may be carried out under a conventional cell culture condition such as at 37°C, in a humidified atmosphere of 5% CO2 in 95% air.
In step 2, the proliferated ES cells are kept in suspension culture with a medium supplemented with LIF and bFGF. Heretofore, LIF has been believed to help retain the ES cells in an undifferentiated state and the art has believed that it is indispensable to exclude LIF from the culture in order to induce differentiation of the ES cells. Accordingly, as far as known to the inventors, all of the proposed EB inducing conditions contain the step culturing the expanded ES cells in suspension culture with a medium containing no LIF to allow their aggregation (for example, Su-Chen Zhang et al., Nature biotechnology, 19, 1129-1133 (2001), the disclosure of the publication is herein incorporated by reference). The present inventors, however, succeeded to provide highly efficient EB formation from the ES cells with a medium comprising LIF and bFGF.
The medium used in step 2 contains LIF and bFGF. The amount of LIF in the medium may preferably be about 100-10000U/ml. The amount of bFGF in the medium may preferably be about 2-100ng/ml. The medium used in this step may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol and nicotinamide, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, laminin, collagen and heparin in a conventional chemically defined cell culture medium. An example of especially preferred medium used in this step is high glucose Dulbecco's modified Eagle's medium (Life Tech.) supplemented with 20% fetal bovine serum replacement (Life Tech.), 2% nonessential amino acid (Life Tech.), 0.1mmol/l 2-mercaptoethanol (Life Tech.), 1000 U/ml of leukemia inhibitor factor (LIF; Life Tech.), 2mmol/l of L- glutamine (Life Tech.) and 4ng/ml of bFGF (R&D systems, Minneapolis).
In step 2, the ES cells are cultured in suspension without the feeder cell layer to allow the cells aggregate to give embryoid bodies. The formation of EBs may be microscopically monitored. According to the present invention, EB formation may be observed from 2 days of the suspension culture. The suspension culture may be continued for 5-10 days to obtain enough amount of EBs. According to the present invention, a significantly larger number of vital EBs are induced than those induced by a conventional suspension culture step with a medium containing no LIF nor bFGF.
The EBs obtained in step 2 are then transferred to a selection-expansion step (step 3). In step 3, thus obtained EBs are plated on a culture container of which inner surface is coated with a protein, such as collagen type IV, and cultured with an appropriate selection-expansion medium. It is preferable to culture the EBs in the protein coated container with the medium used in step 2 for about 2 days and then exchange the medium with a selection- expanding medium.
The selection-expanding medium used in step 3 may preferably be a serum-free cell culture medium supplemented with nicotinamide, insulin and fibronectine. The medium used in this step may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as laminin, collagen and heparin in a conventional chemically defined cell culture medium. An example of preferable medium is a serum free DMEM/F-12 medium supplemented with nicotinamide, fibronectine, and N-2 supplements (GIBCO, 17502-014: consisting of Insulin 500μg/ml, Human transferin 10000μg/ml, Progesterone 0.63μg/ml, Putrescine 1611μg/ml and Selenite 0.52μg/ml in water).
In step 3, the EBs may be cultured with the selection- expanding medium for 3-14 days, preferably for 4-7 days.
The cell clusters obtained in step 3 are then dissociated from the container and plated on a culture container of which inner surface is coated with a protein or an amino acid. The transferred clusters are further cultured in a differentiation medium.
In step 4, the cell clusters can be differentiated into either pancreatic islet like cell clusters or nerve like cells.
In case the islet like cell clusters are desired, the cell clusters may be cultured with a serum-free cell culture medium supplemented with nicotinamide, insulin and laminine. The medium may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, collagen and heparin in a conventional chemically defined cell culture medium. An especially preferred example is serum-free DMEM/F12 medium supplemented with nicotinamide, laminine and N-2 supplement.
According to the present invention, in order to differentiate into nerve like cells, the cell clusters may be cultured with a serum-free cell culture medium supplemented with lysine and laminine. The medium may further comprise one or more growth factors such as activin and nerve growth factor, cytokines such as interleukin-1 and interleukin-2, vitamins such as rethinol and nicotinamide, additional amino-acids such as tyrosine and lysine, and extra cellular matrixes such as fibronectin, collagen and heparin in a conventional chemically defined cell culture medium. An especially preferred example is serum-free DMEM/F12 medium supplemented with lysine, laminine and N-2 supplement.
In step 4, cell clusters may be cultured for 3-90 days or longer. 4-12 days culture will be enough for differentiation into the desired functioning cells and further culture may provide further proliferation of the differentiated clusters.
The pancreatic islet like cell clusters obtained by the present invention represent an ability to produce insulin and to secret insulin in response to glucose stimulation, and the cells consisting the clusters express genes specific to pancreatic endocrine cells including insulin, glucagon, Glut-2 and islet amyloid polypeptide as well as those specific to pancreatic exocrine cells including amylase and carboxypeptitase.
The nerve like cells obtained by the present invention represent nerve fiber like appearances and express markers relevant to nerve cells including nestin, b-tublin III, seletonin, tyrosine hydroxylase. Therefore, said nerve like cells are capable of generating mature neurons.
Since mice ES cells as well as human ES cells proliferate in vitro in an undifferentiated state retaining the pluripotency for more than one year, the present method can be employed to provide enough amount of donor cells used in the cell transplanting therapy.
The present invention further provide a method for treating a mammalian patient having disorders in pancreatic islet function, which comprises transplanting islet-like cell clusters induced from allogenic ES cells according to the invention to the patient. In the present invention,
"mammalian patient having disorders in pancreatic islet function" includes, but not limited to, type I diabetic patient, pancreatomized patient and insulin-required diabetic patient such as type II diabetic patient or patient with cystic fibrosis. The mammalian patient may include human patient. In this embodiment, transplantation of the pancreatic islet like cell clusters obtained as above may be carried out according to a clinically performed or proposed islet transplantation protocol (for example, Kazutomo Inoue and Masaaki Miyamoto, J. Hepatobiliary Pancreat. Surg. 7: 163- 177 (2000), and Wenjing Wang et al., Transplantation 73: 122-129 (2002); the disclosure of the publications are herein incorporated by reference). For example, the pancreatic islet like cell clusters may be implanted intraportally into the liver. Alternatively, the pancreatic islet like cell clusters may be implanted into a prevascularized subcutaneous site. Said clusters may be macroencapsulated with a bio-compatible material before implantation. The amount of the clusters to be transplanted will be determined by the art based on the titer of the obtained clusters as well as the general conditions, age, sex, body weight of the patient to be treated.
Further more, the present invention also provides a method for treating a mammalian patient having disorders in nerve function, which comprises a step of transplanting the nerve like cells derived from allogenic ES cells to the patient. In the present invention, "mammalian patient having disorders in nerve function" includes, but not limited to, patients having nerve degeneration disease such as Alzheimer's disease and Creutzfeldt-Jakob disease or spinal injury. The mammalian patient may include human patient.
The present invention will be further illustrated by the following Examples. The examples are intended to illustrate but not in any means to limit the invention. Example 1
Differentiation of pancreatic islet like clusters from mouse
ES cells
Step 1
Expanding of undifferentiated ES cells In this example, mouse ES cell line 129sv (passages 11; Dainippon Pharmaceutical Co. Ltd., Osaka Japan) was used. A similar study was carried out with mouse ES cell clonal line derived from C57/BL6 mouse (passage 11; kindly provided by Professor Norio NAKATSUJI of Institute for frontier medical sciences, Kyoto University, Kyoto Japan) and similar results as below were obtained (data not shown). Mammalian ES cells can be proliferated in an undifferentiated state if they are cultured on a feeder layer in the presence of leukemia inhibitor factor. Mouse embryo feeder cells (Dainippon Pharmaceutical Co. Ltd., Osaka, Japan) which had been mitotically inactivated with 20 μg/ml mitomycin were used. ES cell culture medium of high glucose Dulbecco's modified Eagle's medium (D-MEM Cat# 12100: Life Technology, Grand, NY) supplemented with 20% fetal bovine serum replacement (Life Tech.), 2% nonessential amino acid (Life Tech.), 0.1mmol/l 2- mercaptoethanol (Life Tech.), 1000 U/ml of leukemia inhibitor factor (LIF; Life Tech.) and 2mmol/l L- glutamine (Life Tech.) was used. A feeder layer of the mitomycin treated mouse embryonic fibroblasts was prepared on a gelatin-coated culture dish (6 cm), 5 ml of the medium was added thereto and 106 of ES cells were plated on the layer. Cells were cultured at 37°C in humidified atmosphere of 5% CO2 in 95% air. Every 3 days, the cells were removed from the dish by means of 0.05% trypsin solution in 0.04% EDTA (Life Tech.) and passaged into a freshly prepared medium on a freshly prepared feeder layer. The ES cells were cultured for 3-7 days. Step 2
Formation of embryoid bodies (EBs)
The ES cells were disassociated by means of the trypsin-EDTA solution and were plated on a non-adherent culture dish to give cell density of 6 x 105 cells/cm2. The cells were kept in suspension culture in the medium used in the above step 1 in the absence or presence of bFGF (4 ng/ml; R&D Systems, Minneapolis, U.S.A. and Kaken Pharmaceuticals, Co. Ltd., Tokyo Japan). Cells were cultured at 37°C in humidified atmosphere of 5% CO2 in 95% air. Every 2 days, the media were replaced with freshly prepared ones.
The cultures were daily observed microscopically. At day 2, the cells cultured with bFGF started to aggregate to generate EBs. The suspension culture was kept for 5 days. At day 5, significantly larger number of cell clusters, i.e. EBs were observed in the culture with bFGF than those previously obtained by the conventional EB inducing process without LIF and bFGF(data not shown). In the group without bFGF, only a few aggregation was observed. Step 3 Selection- Expanding of EBs
The EBs obtained in step 2 with the bFGF containing medium were plated on a Type IV collagen (Sigma, St. Louis, MO) coated 6cm dish filled with the medium used in step 2. After cultured for 48 hr, the medium was replaced with selection-expanding medium of serum free DMEM/F- 12(1:1) medium (cat# 11320, Life Tech.) supplemented with 500 μg/ ml of Bovine Insulin, 1μg / ml of Progestron, 1600 μg /ml of Putrescine and 5 μg / ml of Fibronectin and 10mM of nicotinamide. The cell clusters were cultured for more than 7 days. During the culture, the medium was replaced with freshly prepared one every 3 days. Step 4 Differentiation of the cells After 7 days culture with the selection- expanding medium, further differentiation was induced by culturing the cell clusters with serum free DMEM/F-12(1 :1 ) medium (cat# 11320, Life Tech.) supplenmented with 500 μg/ ml of Bovine Insulin, 1μg/ ml of Progestron, 1600 μg /ml of Putrescine, 1 μg / ml of Laminin and 10mM of nicotinamide. The cells were incubated at 37°C in humidified atmosphere of 5% CO2 in 95% air for 12 days to give islet like cell clusters of about 100-400μm in diameter. RNA extraction and RT- PCR analysis At the end of every step as above, pancreatic relating gene expression on the cells was examined by means of RT-PCR analysis.
Cellular RNA of the cells obtained in each step was. isolated using ISOGEN (Nippon Gene; Osaka, Japan) according to the manufacturer's instruction. The cells were homogenized in 0.8 ml of ISOGEN using a Potter homogenizer at 4°C. The homogenate was mixed with 1 ml of chloroform, and RNA in the aqueous phase was precipitated with the same volume of isopropyl alcohol. Synthesize of cDNA was carried out with oligo dT primers (Takara Shuzo Co. Ltd., Kyoto, Japan) and Moloney murine leukemia virus (M-MLV) Superscript II reverse transcriptase (Gibco/BRL) following the manufacturer's instructions.
Based on thus obtained cDNAs, expression levels of transcription factor mRNAs were determined by means of PCR method. PCR was carried out using standard protocols with Taq polymerase (Boehringer-Mannheim, Indianapolis, IN). Cycling parameters were as follows, denaturation at 94°C for 1min, annealing at 52-61°C for 30- 120 seconds (depending on the primer) for 1min, and elongation at 72°C for 1 min. The number of cycles varied between 25 and 40, depending on the particular mRNA abundance. The number of cycles and the amount of cDNA were chosen in such a way as to select PCR conditions on the linear portion of the reaction curve avoiding "saturation effects" of PCR. Obtained PCR products were confirmed by sequencing.
Primer sequences (forward an reverse), and the length of the amplified products were as follows: β-actin:
ATGGATGACGATATCGCTG
ATGAGGTAGTCTGTCAGGT
569 bp nestin: GGAGTGTCGCTTAGAGGTGC
TCCAGAAAGCCAAGAGAAGC
327 bp i n s u I i n - 1 :
TAGTGACCAGCTATAATCAGAG ACGCCAAGGTCTGAAGGTCC
288 bp insulin-ll:
CCCTGCTGGCCCTGCTCTT
AGGTCTGAAGGTCACCTGCT 212 bp glucagon:
TCATGACGTTTGGCAAGTT
CAGAGGAGAACCCCAGATCA
202 bp Islet Amyloid Polypeptide (IAPP): GATTCCCTATTTGGATCCCC
CTCTCTGTGGCACTGAACCA
221 bp
Glucose transporter 2 (Glut2): CCACCCAGTTTACAAGCTC
TGTAGGCAGTACGGGTCCTC
325 bp
PDX-1:
TGTAGGCAGTACGGGTCCTC CCACCCCAGTTTACAAGCTC
325 bp amylase-2A
CATTGTTGCACCTTGTCACC
TTCTGCTGCTTTCCCTCATT 300 bp carboxypeptidase A:
GCAAATGTGTGTTTGATGCC
ATGACCAAACTCTTGGACCG
521 bp GATA-4:
CGCCGCCTGTCCGCTTCC
TTGGGCTTCCGTTTTCTGGTTTGA
193 bp
HNF3: ACCTGAGTCCGAGTCTGACC GGCACCTTGAGAAAGCAGTC
345 bp
OCT-4:
GGCGTTCTCTTTGGAAAGGTGTTC
CTCGAACCACATCCTTCTCT
293bp
Results are shown in table 1 below;
Table 1
Gene expression on the cells cultured in the presence of bFGF in step2
Figure imgf000028_0001
The gene of pancreatic transcription factor PDX-1, which is indispensable for pancreatic development, was expressed in steps 1 and 4 cells. Oct-4, which relates to differentiation of ES cell, was expressed in step 1 cells and down- regulated with the differentiation of the ES cells. The ES cells at every step expressed a maker of definitive (embryonic) and visceral (extra-embryonic) endoderm GATA-4 and definitive endoderm HNF3β concerning markers of pancreatic βcell fate. Nestin, a transcription factor relates to immature hormone-negative pancreatic cells, was strongly expressed in step 2 and down-regulated with the differentiation of the ES cells. The results showed that many nestin positive progenitor cells were contained in the EBs obtained in the presence of bFGF. Interestingly, EBs induced by bFGF treatment expressed transcription factors of endocrine (Insulin I, Insulin II, Glucagon, Glucose transporter-2 (Glut-2) and Islet Amyloid Polypeptide) specific genes whereas any gene concerning pancreatic islet cells did not expressed in the cell clusters obtained in step 2 using the medium without bFGF. In steps 3 and 4, the cells expressed exocrine specific genes (amylase and carboxypeptidase). These results indicates that pancreatic islet like cell clusters of the invention can be matured to a pancreatic tissue structure, which composed of endocrine cells including glucagon-producing α cells, insulin- producing β cells, pancreatic polypeptide-producing y cells, and somatostin-producing δ cells and exocrine cells. Insulin Secretion Test
The cell clusters obtained in step 4 (20-25 clusters) were washed 3 times with PBS(-) and plated on a 6cm cell culture dish containing Krebs-Ringer with bicarbonate buffer consisting of 120 mM NaCl, 5 mM KCI, 2.5 mM CaCI2, 1.1 mM MgCI2, 25 mM NaHC03 and 0.1% bovine serum albumin, and incubated at 37°C. 3.3 mmol/l (L) or 25mmoi/l (H) glucose was added thereto and incubated. Five and thirty minutes after the glucose stimulation, the insulin contents in the buffer were measured using insulin enzyme- linked immunosorbence assay (ELISA) kit (ALPCO, Windham, NH). Results are shown in Figure 2. In the Fig. 2, the amounts of insulin secreted per one cluster in response to the low or high glucose stimulation at 5 and 30 minutes after the stimulation are shown. The clusters exhibited insulin secretion in response to glucose stimulation in a dose dependent manner.
For determination of total cellular insulin content, the cell clusters obtained in step 4 were extracted with acid ethanol (10% glacial acetic acid in absolute ethanol) overnight at 4°C, followed by cell sonication and then, the insulin content in the supernatant was determined by means of the ELIZA kit. Total cellular protein amount was determined using DC protein assay system (Bio-Rad laboratories, Hercules, CA). The total cellular insulin content of those cell clusters was 71.3ng/mg protein. Histological and Immunohistochemistry Analysis
Paraffin slices of the cell clusters obtained in step 4 were prepared as follows. The cell clusters (in step 4, incubated 12 days) were washed three times with ice-cold PBS and were fixed with methanol/aceton (1:1) for over night. The clusters were dehydrated with aqueous alcohol (70-100%), then embedded in a paraffin block and the block was sliced to give 4μm and 8μm thick slices.
Thus obtained 4μm thick slices were histologically evaluated with hematoxylin/eosin staining.
In order to immunohistochemical evaluation, 8μm thick slices were stained with antibodies by means of the standard protocol. Primary antibodies used herein were follows: nestin rabbit polyclonal 1:500 (Dako, Carpinteria, CA), tubulin type III (TuJ1) mouse monoclonal 1:500 (Babco, Richmond, CA), tubulin type III (TuJ1) rabbit polyclonal 1:2000 (Babco, Richmond, CA), insulin mouse monoclonal 1:1000 (Sigma, St. Louis, MO), insulin guinea pig polyclonal 1:100 (DAKO, Carpinteria, CA), glucagon rabbit polyclonal (DAKO, Carpinteria, CA).
In order to detect the primary antibodies, fluorescently labeled secondary antibodies (Jackson Immunoresearch Laboratories, West Grove, PA) were used according to the supplier's instruction.
The obtained insulin producing cell cluster was strongly positive to insulin and glucagone, and positive to nestin and TuJ1. Example 2 Transplantation of the insulin producing cell clusters into STZ derived diabetic mice.
The insulin producing pancreatic islet like cell clusters were transplanted to determine if the cluster could differentiate into functioning pancreatic islet in vivo. All animal studies were carried out in accordance with
Guideline for Animal Experiments of Kyoto University. Experimental diabetic mice were prepared according to the method disclosed in H. Iwata et al., Pancreas vol. 23(4) 375-381(2001), the disclosure of the publication is herein incorporated by reference. Streptozotocin (STZ) cryopreserved powder (Sigma, St. Louis, MO) was dissolved in 0.1 M citrate buffer, pH 4.5 before use. The STZ solution was intraperitoneally injected (227 mg/kg of body weight) to 8- to 10-weeks-old male Nude mice (Shimizu, Kyoto, Japan), Stable hyperglycemia, i.e. increased blood glucose levels of about 350 - 600 mg/dl) were usually developed 7 to 10 days after the STZ single injection.
Blood glucose level of the mouse was determined using Glucometer Elite. XL blood glucose meter (Fujii Corp., Tokyo, Japan). Animals represent 350mg/dl or more non- fasting blood glucose at 7-10 days of STZ injection were regarded as diabetic mice and used at 14 day from the STZ injection.
14 days after the STZ injection, the diabetic animals were grafted with 3000 insulin producing pancreatic islet like cell clusters obtained in Example 1 or received sham operation. Under nembutal anesthetization, the cell clusters suspended in PBS(-) were injected into the kidney subcapsuler region (one kidney) of the diabetic mice with 23-gauge winged needle. For the sham-operating group, the same volume of PBS(-) was injected in the same manner as above. The experimental group received cell clusters, non-treated control group and sham group consisted of 6, 3 and 3 animals respectively. After the transplantation, non-fasting blood glucose and body weight were monitored daily. The results are shown in Figures 3 and 4.
One day after the transplantation, the blood glucose of the experimental group significantly decreased and the significantly lower blood glucose level than the sham group was kept throughout the time of the experiment. The body weight of the implanted group increased slightly and kept stable.
At days 14 and 21, 2 and 4 animals implanted with the clusters were sacrificed respectively. All implanted mice remained healthy until killed and kept significantly lower blood glucose than the sham group. To the contrarily, blood glucose levels in non-treated control and sham groups were increased gradually and became exhausted. All of the mice of control and sham groups died prematurely from complication of diabetics between day 14 and day 30 of the operation.
From the sacrificed animals, the implanted tissue was excised, fixed with 4% paraformaldehyde in PBS and embedded in paraffin block. Thus obtained tissue slices of 4-8μm thickness were immunohistochemically examined in the same manner as Example 1.
At the implanted region, single massed endocrine cells which were immunohistochemically positive to insulin and glucagone were observed. There was no teratoma observed at the area. Example 3 Induction of nerve like cells
The mouse ES cells same as used in Example 1 were treated in the same manner as steps 1-3 of Example 1. Thus obtained cell clusters were then cultured in a dish coated with poly-L-lysine and filled with serum free DMEM/ F-12(1:1) medium (cat# 11320, Life Tech.) supplemented with 500 μg/ ml of Bovine Insulin, 1μg/ ml of Progestron, 1600 μg /ml of Putrescine, 10mM of lysine and 1μg / ml of Laminin. The cells were cultured for 12 days and the obtained cells were examined genetically and immunohistochemically according to the same manner as described in Example 1. In this example, thyrosine hydroxylase(TH) polyclonal 1:200 (Pel-Freeze, Rogers, AR), thyrosine hydroxylase(TH) monoclonal 1:1000 (Sigma, St. Louis, MO), serotonin polyclonal 1:4000 (Sigma, St. Louis, MO), MAP 2 polyclonal (Chemicon International, Temecula, CA), and GFAP monoclonal (Clon Tech, Palo Alto, CA) were used in addition to the antibodies used in Example 1.
The obtained cells represented nerve fiber like appearance and were immunohistochemically positive to nestin, TuJ1 (β-tublin III), serotonin, GFAP, MAP 2 and tyrosine-hydroxylase. By means of RT-PCR described in Example 1, the nerve like cells was confirmed to express Nurt-1 transcription factor. The primer sequences used herein for detecting Nurt-1 were as follows: TGAAGAGAGC GGAGAAGGAG ATC TCTGGAGTTA AGAAATCGGA GCTG 255 bp.
Accordingly, the obtained nerve like cells are capable of generating mature neurons if they are implanted in vivo. Example 4 Human ES cells
According to the same manner as described in Example 1, insulin producing pancreatic islet like cell clusters are obtained from human ES cells. ES cells may be those described in the art such as U.S. patent Nos. 5,843,780 and 6,200,806; Science 282, 1145-1147 (1998), the disclosures of the publications are herein incorporated by reference. Thus obtained cell clusters produce insulin and secret insulin in response to glucose in a dose dependent manner. The cell clusters are implanted into a human type I diabetic patient. About 3-5 105 clusters are suspended in about 50-100 ml of Krebs-Ringer solution and the suspension is injected into the liver via portal vein, or is implanted to subcutaneous space as a bio-artificial pancreas. The pancreatic function of the implanted patient restores and the patient acquires insulin-independency. Example 5
Induction of dopaminergic (DA) neuron from Mouse embryonic stem cells
Basically in the same manner as Example 1, differentiation of Mouse ES cell line 129sv (Dainippon- pharm. Co. LTD., Osaka, Japan) was induced. Step 1
The cells were propagated on a feeder layer of mitomycin treated mouse embryonic fibroblasts in DMEM (Invitrogen, Grand Island, NY, USA) medium containing 20% fetal bovine serum (FBS, JRH Bioscience, Lenexa, KS, USA), 1000 unit/ml leukimia inhibitory factor (LIF), 50mM 2- mercaptoethanol, 2mM L-glutamic acid and 10X non- essential amino acid (Invitrogen, Grand Island, NY, USA). ES cell colonies were detached and subcultured until a desired amount of the cells is obtained. Step 2
To induce EB formation, single-cells disassociated by trypsin-EDTA were plated onto nonadherent bacterial dishes at a density less than 6 x 105 cells/cm2 and EBs were kept in suspension culture for 3 days in the medium described above in the presence of bFGF (4 ng/ml; R&D Systems, Minneapolis, USA). Step 3 After the EBs were formed, they were plated onto Poly
(L-lysine)-coated dishes (Falcon Labware, Bedford, MA) in the presence of EB culture medium used in the above step. After cultured for 24 hr, the EB culture medium was replaced with serum free DMEM/F-12 (Invitrogen, Grand Island, NY^ USA) medium containing 500 μ g/ml of bovine insulin, 1 μ g/ml of progestron, 1600 μ g/ml of putrescine, 0, 5 or 10mM of nicotinamide and 5 μ gl \ of fibronectin (Nakalai tesque, Kyoto), and cultured for 4 days. Step 4 After 4 days of culture, cell differentiation were induced by replacing the medium with serum free DMEM/F- 12 medium containing 500 μ gl ml of bovine insulin, 1 μ g/ml of progestron, 1600 μ g/ml of putrescine, 0, 5 or 10mM of nicotinamide, 5 μ gl ml of fibronectin (Nakalai tesque, Kyoto, Japan) and 1 μ gl ml of laminin (Nakalai tesque, Kyoto, Japan). The cells were cultured and obtained cells were examined as follows.
TuJ-positive colonies
Colonies obtained as above were subjected to immunohistochemical analysis for expression of the neuronal marker TuJ. Results are shown in Fig. 5.
In order to examine the effect of bFGF in step 2, TuJ- expression on the colonies obtained from the ES cells which were treated as above except for cultured in the presence (4 ng/ml) or absence of bFGF in step 2, and then cultured in the presence of 10 mM of nicotineamide in steps
3 and 4 were examined. Results are shown in Fig. 6. In
Fig. 6, those cultured in the presence or absence of bFGF are indicated as "treated" or "untreated" respectively. These results demonstrate that both NA and bFGF enhance the number of colonies that express TuJ.
Expression of neural relating gene on the cells
By means of RT-PCR described in Example 1, the undifferentiated ES cells (step 1) and those obtained in step 4 were genetically examined. The examined cells were those treated with bFGF in step 2 and cultured in the presence of 10mM NA in steps 3 and 4. The primers used in this examination are follows:
Nurt-1: tyrosin hydroxylase TGAAGAGAGC GGAGAAGGAG ATC TCTGGAGTTA AGAAATCGGA GCTG
Wnt-1: Mesencephalic domaminergic neuron transcription factor signaling molecule
5*-ACCTGTTGACGGATTCCAAG-3' 5*-TCATGAGGAAGCGTAGGTCC-3';
Engrailed-1 (En 1): Mesencephalic domaminergic neuron transcription factor
5'-TCAAGACTGACTCACAGCAACCCC-3'
5'-CTTTGTCCTGAACCGTGGTGGTAG-3' Ptc: SHH signal receptor, patched
5'-CCTCCTTTACGGTGGACAAA-3'
5'-ATCAACTCCTCCTGCCAATG-3'
Glycoprotein sonic hedgehog (Shh)
5'-GGAAGATCACAAGAAACTCCGAAC-3' 5'-GGATGCGAGCTTTGGATTCATAG-3'
Otx 1: Transcription factor critical in brain morphogenesis
5'-GCTGTTCGCAAAGACTCGCTAC-3'
5'-CCATGACCTATACTCAGGCTTCAGG-3';
Otx 2: Transcription factor critical in brain morphogenesis δ'-CCATGACCTATACTCAGGCTTCAGG-S'
5'-GAAGCTCCATATCCCTGGGTGGAAAG-3'
Pax 2:Mesencephalic domaminergic neuron transcription factor
5'-CCAAAGTGGTGGACAAGATTGCC-3' 5'-GGGATAGGAAGGACGCTCAAAGAC-3' Pax 5:Mesencephalic domaminergic neuron transcription factor
5'-CAGATGTAGTCCGCCAAAGGATAG-3' δ'-ATGCCACTGATGGAGTATGAGGAGCC-S'
Smo: SHH signal receptor, smoothened
5'-CTGAGAGTGCCAGAAAAGGG-3'
5'-TCATCATGCTGGAGAACTCG-3'
Fibroblast growth factor receptor (FGFR 3R)
5'-ATCCTCGGGAGATGACGAAGAC-3'
5'-GGATGCTGCCAAACTTTGTTCTC-3'
Results are shown in table 2
TABLE 2
Figure imgf000040_0001
As is shown in table 2, colonies induced according to the instant invention expressed high level of neural specific genes encoding transcription factors and signaling molecules. Development of neural network like appearance
The obtained colonies were microscopically examined to determine the degree of complexities of the neural network like appearance. Results are shown in Fig. 7. Fig. 7 is histogram summarizing the effect of NA (nicotineamide) and bFGF on complexities of the neural network like appearance of the EBs. The EBs were cultured in the presence or absence of 4 ng/ml of bFGF in step 2 (indicated "non-treated" or "treated" respectively) and then cultured in the presence of NA in steps 3 and 4. Each condition was assayed by counting all EBs in culture and the experiment was repeated twice resulting in similar results. BMP-4 or FBS treatment The cells treated with bFGF in step 2 were then cultured in the presence of 0.5 nM bone morphogenetic protein (BMP-4) or 5% fetal bovine serum (FBS) in addition to 10mM of NA in the following steps (steps 3 and 4). In this procedure, BMP4 was freshly added at each medium change. Both BMP-4 and FBS suppress differentiation of the cells into neural cells.
Results are shown in Fig. 8. Both of BMP-4 and FBS suppressed the expression of TuJ on the colonies. Effects of transplantation of the differentiated cells on 6- OHDA lesion and rotational behavior
It is considered that a unilateral dopaminergic neurogenerative animal induced by 6-OHDA is a useful model of Parkinson's disease (PD), since loss of nigral dopaminergic neurons is characterized in PD pathology. It is known that marked dopamine depletion (over 95%) in the striatum induces the supersensitivity in dopamine receptors. In 6-OHDA-lesioned rats, therefore, a mixed D1/D2 receptor agonist apomorphne induces rotational asymmetry in the contralateral direction, while a DA releaser amphetamine induces the ipsilateral rotation. Accordingly, we studied in vivo effects of transplantation of the differentiated and undifferentiated ES cells on the 6-OHDA lesion model.
All animal studies were in accordance with Kyoto University guidelines and the National Institutes of Health Guide for the Care and Use of Laboratory Animals. Experimental animal used in 8- week-old male Wister rats, weighing approximately 280g (SLC Inc., Hamamatsu, Japaη). The rats were fasted overnight with free access to water. 6- Hydroxydpamine (6-OHDA) solution was prepared by dissolving 12g of 6-OHDA (Sigma, St Louis, MO, USA), in a final volume of 41 sterilized physiological saline containing 0.02% ascorbic acid. For stereotaxic microinjection, rats were anesthetized (sodium pentobarbital, 50 mg/kg, i.p.) and immobilized in Kopf stereotaxic frame. Subsequently, the rats were injected with the 6-OHDA solution into left substantia nigra, where the bregma was -4.8 mm caudal, 1.8 mm left lateral, -7.8 mm ventral, via a motor-driven 10 μl-Hamilton syringe using 26 gauge needle. Coordinates were set according to the rat brain atlas (Paxinos G. and Watson C. (1986) "The rat brain in stereotaxic coordinates" (Second Edition). Academic Press, North Ryde, Australia). After 2 weeks, lesioned rats were selected for transplantation by estimation of rotational behavior in response to apomorphine (Sigma). The rotational testing was performed in the rotometer bowls (Ungerstedt, U. (1971) "Postsynaotic supersensitivity after 6- hydroxydopamine induced degeneration of the nigro-striatal dopamine system" Acta Physiol. Scand., 82 Suppl. 367, 69- 93) and the total number of full 360° rotations in contralateral directions was counted. Animals that showed a strong contralateral rotational behavior induced by an administration of apomorphine (0.6 mg/kg, i.p.) were selected for the following transplantation surgery. Transplantation surgery 6-OHDA-lesioned rats were injected with the vehicle
(sterilized phosphate-buffered saline (PBS) n = 10, a:vehicle) or cell suspension containing low numbers of undifferentiated ES cells (about 4000 cells of ES cells from step 1, n = 10, b:ES-L), and differentiated cells (about 20,000 cells of the cells from step 4 day 4, n = 10, i:4F).
The selected rats were anesthetized (sodium pentobarbital, 50 mg/kg, i.p.) and then placed in Kopf stereotaxic frame. Each animal received a microinjection of 1.0 μ l of grafting cell suspension or the vehicle into two sites of the left striatum, where the bregma was +1.0 mm caudal, 3.0 mm left later, -5.5 and 5.0 mm ventral according to the rat brain atlas (id), using a 10 μ l-Hamilton syringe with 22 gauge needle. A 5-min waiting period allowed transplanted cells to settle before the needle was removed. Subsequently, apomorphine-induced rotational asymmetry was assesses every 2 weeks.
The number of full 360° rotations in the contralateral direction was counted for 60 min after administration of apomorphine (0.6 mg/kg, i.p.). Statistics
Results are shown in Figs. 9-11. Data are presented as mean±SEM and compared using the unpaired Student's test or one-way analysis of variance. Differences were considered significant at P<0.05. Further statistical analysis for post hoc comparison was performed using the Bonferroni/Dunn test (Statview, Abacus Concepts, Berkely, USA) for the numbers of apomorphine-induced rotational asymmetry.
Apomorphine-induced rotation asymmetry was significantly improved by transplantation of differentiated DA-neurons in step 4 after 2 weeks. However, other transplanted cells did not efficiently contralateral rotation. Significance (Bonferroni/Dunn post hoc comparisons after ANOVA): *p<0.05, **p<0.01, ***p<0.001 vs. each time point in the vehicle-injection (a); ! p<0.05, !!! p<0.001 vs. pre- transplantation in each group.
In this study, undifferentiated mES cells and the vehicle did not improve apomorphne induced rotational asymmetry. In contrast, transplantation of the ES cells treated according to the instant invention (at step 4) significantly reduced the apomorphine induced rotational asymmetry after 2 weeks. In addion, this restoration sustained until completed behavioral assessment at 12 weeks. According to the results of the example 5, it can be concluded that mouse ES cells were differentiated into dopaminergic nerve cells according to the method of the present invention.
According to the same manner as described in the above examples, nerve like cell can be obtained from human ES cells. Human ES cells may be those described in Example 4. Although some minor modifications would be required for optimizing the culture conditions, it would be mere routine work for the art. According to he present invention, dopaminergic neurons can effectively enriched from the total population of differentiated ES cells without gene modification, flow cytometry sorting or separation with magnetic beads. The nerve like cells induced from ES cells by the simple 4-step method of the instant invention represent good therapeutic effect on Perkinson's disease and the patients received the treatment can survive long- term .

Claims

1. A method for inducing differentiation of mammalian embryonic stem cells into functioning cells, which comprises the steps of; 1) culturing the mammalian embryonic stem cells together with feeder cells with a medium comprising leukemia inhibitor factor;
2) culturing the obtained cells in absence of feeder cells with a medium comprising leukemia Inhibitor factor and basic FGF in a suspension culture condition to give embryonic bodies;
3) culturing the obtained embryonic bodies with a selection-expanding medium; and
4) culturing the obtained cell clusters with a differentiation medium to give functioning cells.
2. The method of claim 1, wherein the medium used in step 2) comprises about 100-10000 U/ml of leukemia inhibitor factor.
3. The method of claim 1, wherein the medium used in step 2) comprises about 2-100 ng/ml of bFGF.
4. The method of claim 1, wherein the medium used in step 3) comprises nicotinamide, insulin and fibronectine in an serum-free cell culture medium.
5. The method of claim 1, wherein the functioning cells are insulin producing pancreatic islet like cell clusters.
6. The method of claim 5 wherein the medium used in step 4) comprises nicotinamide, insulin and laminine in a serum-free cell culture medium.
7. The method of claim 1, wherein the functioning cells are nerve like cells.
8. The method of claim 7 wherein the medium used in step 4) comprises L-lysine, insulin and laminine in a serum- free cell culture medium.
9. Functioning cells induced from mammalian ES cells by the method of claim 1.
10. Insulin secreting cell clusters induced from mammalian ES cells by the method of claim 5.
11. Nerve like cells induced from mammalian ES cells by the method of claim 7.
12. A method for treating a mammalian patient having disorders in pancreatic islet function, which comprises implanting pancreatic islet-like cell clusters induced from allogenic ES cells by the method of claim 5 to the patient.
13. The method of claim 12, wherein the patient is type I diabetic patient.
14. A method for treating a mammalian patient having disorders in nerve function, which comprises implanting nerve like cells induced from allogenic ES cells by the method of claim 7 to the patient.
PCT/JP2003/000699 2002-01-25 2003-01-27 Method for inducing differentiation of embryonic stem cells WO2003062405A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2003562273A JP2005514944A (en) 2002-01-25 2003-01-27 Method for inducing differentiation of embryonic stem cells into functional cells

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/054,789 2002-01-25
US10/054,789 US20030162290A1 (en) 2002-01-25 2002-01-25 Method for inducing differentiation of embryonic stem cells into functioning cells

Publications (2)

Publication Number Publication Date
WO2003062405A2 true WO2003062405A2 (en) 2003-07-31
WO2003062405A3 WO2003062405A3 (en) 2003-10-16

Family

ID=27609146

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2003/000699 WO2003062405A2 (en) 2002-01-25 2003-01-27 Method for inducing differentiation of embryonic stem cells

Country Status (3)

Country Link
US (2) US20030162290A1 (en)
JP (1) JP2005514944A (en)
WO (1) WO2003062405A2 (en)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004099395A2 (en) * 2003-05-08 2004-11-18 Cellartis Ab A method for the generation of neural progenitor cells
WO2005123902A1 (en) * 2004-06-18 2005-12-29 Riken Method of inducing the differentiation of embryonic stem cells into nerve by serum-free suspension culture
JP2006218035A (en) * 2005-02-09 2006-08-24 Tokyo Univ Of Agriculture & Technology Method of producing myocardial graft and myocardial differentiation accelerator
WO2006060753A3 (en) * 2004-12-03 2006-11-16 Rhode Island Hospital Diagnosis and treatment of alzheimer's disease
EP1783208A1 (en) * 2004-07-06 2007-05-09 Kyowa Hakko Kogyo Co., Ltd. Method of producing nerve cell
CN100389194C (en) * 2004-07-16 2008-05-21 ***昆明总医院 Insulin secretory cell extract and its application in inducing stem cell differentiation
US7638328B2 (en) 2003-05-08 2009-12-29 Cellartis Ab Method for efficient transfer of human blastocyst-derived stem cells (hBS cells) from a feeder-supported to a feeder-free culture system, long-term propagation of hBS cells under feeder-free conditions and use of cultured hBS cells for applications in myocardial regeneration
US7833513B2 (en) 2004-12-03 2010-11-16 Rhode Island Hospital Treatment of Alzheimer's Disease
EP2302036A2 (en) 2005-05-27 2011-03-30 Lifescan, Inc. Amniotic fluid derived cells
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US8741643B2 (en) 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
US8778673B2 (en) 2004-12-17 2014-07-15 Lifescan, Inc. Seeding cells on porous supports
US8785185B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US8785184B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9012218B2 (en) 2008-10-31 2015-04-21 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9062290B2 (en) 2007-11-27 2015-06-23 Lifescan, Inc. Differentiation of human embryonic stem cells
US9074189B2 (en) 2005-06-08 2015-07-07 Janssen Biotech, Inc. Cellular therapy for ocular degeneration
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
US9096832B2 (en) 2007-07-31 2015-08-04 Lifescan, Inc. Differentiation of human embryonic stem cells
US9133439B2 (en) 2009-12-23 2015-09-15 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9150833B2 (en) 2009-12-23 2015-10-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
JP2015180215A (en) * 2004-04-27 2015-10-15 ヴィアサイト,インコーポレイテッド PDX1 expressing endoderm
US9181528B2 (en) 2010-08-31 2015-11-10 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9234178B2 (en) 2008-10-31 2016-01-12 Janssen Biotech, Inc. Differentiation of human pluripotent stem cells
US9434920B2 (en) 2012-03-07 2016-09-06 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
US9506036B2 (en) 2010-08-31 2016-11-29 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9528090B2 (en) 2010-08-31 2016-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9593305B2 (en) 2008-06-30 2017-03-14 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9752125B2 (en) 2010-05-12 2017-09-05 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9969973B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Methods and compositions for cell attachment and cultivation on planar substrates
US9969972B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Pluripotent stem cell culture on micro-carriers
US9969981B2 (en) 2010-03-01 2018-05-15 Janssen Biotech, Inc. Methods for purifying cells derived from pluripotent stem cells
US10006006B2 (en) 2014-05-16 2018-06-26 Janssen Biotech, Inc. Use of small molecules to enhance MAFA expression in pancreatic endocrine cells
US10066203B2 (en) 2008-02-21 2018-09-04 Janssen Biotech Inc. Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US10066210B2 (en) 2012-06-08 2018-09-04 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
US10076544B2 (en) 2009-07-20 2018-09-18 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US10138465B2 (en) 2012-12-31 2018-11-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using HB9 regulators
US10344264B2 (en) 2012-12-31 2019-07-09 Janssen Biotech, Inc. Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
US10358628B2 (en) 2011-12-22 2019-07-23 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into single hormonal insulin positive cells
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
US10377989B2 (en) 2012-12-31 2019-08-13 Janssen Biotech, Inc. Methods for suspension cultures of human pluripotent stem cells
US10421942B2 (en) 2003-12-23 2019-09-24 Viacyte, Inc. Definitive endoderm
US10420803B2 (en) 2016-04-14 2019-09-24 Janssen Biotech, Inc. Differentiation of pluripotent stem cells to intestinal midgut endoderm cells
US10465162B2 (en) 2004-04-27 2019-11-05 Viacyte, Inc. Anterior endoderm cells and methods of production

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003224681A1 (en) * 2002-03-13 2003-09-29 Bresagen Inc. Compositions and methods for primate neural cell production
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
WO2005038012A2 (en) * 2003-06-27 2005-04-28 Ethicon Incorporated Cartilage and bone repair and regeneration using postpartum-derived cells
US20050060166A1 (en) * 2003-09-15 2005-03-17 Durham Jon A. Method of providing funeral products and services
US20060171930A1 (en) * 2004-12-21 2006-08-03 Agnieszka Seyda Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
CA2589063C (en) 2004-12-23 2016-08-09 Ethicon Incorporated Treatment of parkinson's disease and related disorders using postpartum derived cells
PL1831356T3 (en) 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20060183712A1 (en) * 2005-02-17 2006-08-17 The Texas A&M University System Affinity purified heparin/heparan sulfate for controlling the biological activity of the FGF receptor
WO2007070870A1 (en) 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
EP1976975B1 (en) 2005-12-19 2012-08-01 Ethicon, Inc. In vitro expansion of postpartum derived cells in roller bottles
EP1979050B1 (en) * 2005-12-28 2017-04-19 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using postpartum-derived cells
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
JP2007267672A (en) * 2006-03-31 2007-10-18 Nippon Menaade Keshohin Kk Differentiation inducting method in stem cell of mammal
UA99152C2 (en) 2007-10-05 2012-07-25 Этикон, Инкорпорейтед Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
BRPI0923070A2 (en) * 2008-12-19 2016-06-14 Atrm Llc "Uses of compositions for regeneration and repair of neural tissue after injury, said compositions, and kit"
PL2379088T3 (en) 2008-12-19 2018-07-31 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
SI2379087T1 (en) * 2008-12-19 2015-01-30 DePuy Synthes Products, LLC Umbilical cord tissue derived cells for treating neuropathic pain and spasticity
AU2010229651B2 (en) 2009-03-26 2014-05-08 Advanced Technologies And Regenerative Medicine, Llc Human umbilical cord tissue cells as therapy for Alzheimer' s disease
WO2012009830A1 (en) 2010-07-22 2012-01-26 宁夏医科大学附属医院 Methods for producing nerve cells from stem cells, nerve cells and uses thereof
KR101981450B1 (en) 2011-12-23 2019-08-28 디퍼이 신테스 프로덕츠, 인코포레이티드 Detection of human umbilical cord tissue-derived cells
AU2014364632B2 (en) * 2013-12-16 2021-04-01 Fresenius Medical Care Deutschland G.M.B.H. Pancreatic islet-like cell structures and a method of preparing thereof
JP6822668B2 (en) * 2015-10-28 2021-01-27 学校法人近畿大学 Method for Embryoid Body Formation of Pluripotent Stem Cell and Composition for Embryoid Body Formation of Pluripotent Stem Cell

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5690926A (en) * 1992-10-08 1997-11-25 Vanderbilt University Pluripotential embryonic cells and methods of making same
WO2001083715A2 (en) * 2000-05-01 2001-11-08 THE GOVERNMENT OF THE UNITED STATES OF AMERICA as represented by the Secretary, Derivation of midbrain dopaminergic neurons from embryonic stem cells

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6331406B1 (en) * 1997-03-31 2001-12-18 The John Hopkins University School Of Medicine Human enbryonic germ cell and methods of use
US6090622A (en) * 1997-03-31 2000-07-18 The Johns Hopkins School Of Medicine Human embryonic pluripotent germ cells
US6602711B1 (en) * 2000-02-21 2003-08-05 Wisconsin Alumni Research Foundation Method of making embryoid bodies from primate embryonic stem cells

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5690926A (en) * 1992-10-08 1997-11-25 Vanderbilt University Pluripotential embryonic cells and methods of making same
WO2001083715A2 (en) * 2000-05-01 2001-11-08 THE GOVERNMENT OF THE UNITED STATES OF AMERICA as represented by the Secretary, Derivation of midbrain dopaminergic neurons from embryonic stem cells

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LUMELSKY NADYA ET AL: "Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets" SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE,, US, vol. 292, no. 5520, 2001, pages 1389-1394, XP002183377 ISSN: 0036-8075 *
OTONKOSKI T ET AL: "NICOTINAMIDE IS A POTENT INDUCER OF ENDOCRINE DIFFERENTIATION IN CULTURED WOMEN FETAL PANCREATIC CELLS" JOURNAL OF CLINICAL INVESTIGATION, NEW YORK, NY, US, vol. 92, no. 3, September 1993 (1993-09), pages 1459-1466, XP009008891 ISSN: 0021-9738 *
SCHULDINER MAYA ET AL: "Effects of eight growth factors on the differentiation of cells derived from human embryonic stem cells" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE. WASHINGTON, US, vol. 97, no. 21, 10 October 2000 (2000-10-10), pages 11307-11312, XP002184277 ISSN: 0027-8424 *

Cited By (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7638328B2 (en) 2003-05-08 2009-12-29 Cellartis Ab Method for efficient transfer of human blastocyst-derived stem cells (hBS cells) from a feeder-supported to a feeder-free culture system, long-term propagation of hBS cells under feeder-free conditions and use of cultured hBS cells for applications in myocardial regeneration
WO2004099395A3 (en) * 2003-05-08 2005-03-17 Cellartis Ab A method for the generation of neural progenitor cells
WO2004099395A2 (en) * 2003-05-08 2004-11-18 Cellartis Ab A method for the generation of neural progenitor cells
US10421942B2 (en) 2003-12-23 2019-09-24 Viacyte, Inc. Definitive endoderm
US10465162B2 (en) 2004-04-27 2019-11-05 Viacyte, Inc. Anterior endoderm cells and methods of production
JP2015180215A (en) * 2004-04-27 2015-10-15 ヴィアサイト,インコーポレイテッド PDX1 expressing endoderm
US11746323B2 (en) 2004-04-27 2023-09-05 Viacyte, Inc. PDX1 positive foregut endoderm cells and methods of production
US8492147B2 (en) 2004-06-18 2013-07-23 Riken Method of inducing the differentiation of embryonic stem cells into nerve by serum-free suspension culture
WO2005123902A1 (en) * 2004-06-18 2005-12-29 Riken Method of inducing the differentiation of embryonic stem cells into nerve by serum-free suspension culture
EP1783208A4 (en) * 2004-07-06 2007-11-14 Kyowa Hakko Kogyo Kk Method of producing nerve cell
EP1783208A1 (en) * 2004-07-06 2007-05-09 Kyowa Hakko Kogyo Co., Ltd. Method of producing nerve cell
CN100389194C (en) * 2004-07-16 2008-05-21 ***昆明总医院 Insulin secretory cell extract and its application in inducing stem cell differentiation
US7833513B2 (en) 2004-12-03 2010-11-16 Rhode Island Hospital Treatment of Alzheimer's Disease
WO2006060753A3 (en) * 2004-12-03 2006-11-16 Rhode Island Hospital Diagnosis and treatment of alzheimer's disease
US8778673B2 (en) 2004-12-17 2014-07-15 Lifescan, Inc. Seeding cells on porous supports
JP2006218035A (en) * 2005-02-09 2006-08-24 Tokyo Univ Of Agriculture & Technology Method of producing myocardial graft and myocardial differentiation accelerator
EP2302036A2 (en) 2005-05-27 2011-03-30 Lifescan, Inc. Amniotic fluid derived cells
US9074189B2 (en) 2005-06-08 2015-07-07 Janssen Biotech, Inc. Cellular therapy for ocular degeneration
US8741643B2 (en) 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
US9725699B2 (en) 2006-04-28 2017-08-08 Lifescan, Inc. Differentiation of human embryonic stem cells
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
US10316293B2 (en) 2007-07-01 2019-06-11 Janssen Biotech, Inc. Methods for producing single pluripotent stem cells and differentiation thereof
US10456424B2 (en) 2007-07-31 2019-10-29 Janssen Biotech, Inc. Pancreatic endocrine cells and methods thereof
US9744195B2 (en) 2007-07-31 2017-08-29 Lifescan, Inc. Differentiation of human embryonic stem cells
US9096832B2 (en) 2007-07-31 2015-08-04 Lifescan, Inc. Differentiation of human embryonic stem cells
US9062290B2 (en) 2007-11-27 2015-06-23 Lifescan, Inc. Differentiation of human embryonic stem cells
US9969982B2 (en) 2007-11-27 2018-05-15 Lifescan, Inc. Differentiation of human embryonic stem cells
US11001802B2 (en) 2008-02-21 2021-05-11 Nunc A/S Surface of a vessel with polystyrene, nitrogen, oxygen and a static sessile contact angle for attachment and cultivation of cells
US10066203B2 (en) 2008-02-21 2018-09-04 Janssen Biotech Inc. Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US9845460B2 (en) 2008-04-24 2017-12-19 Janssen Biotech, Inc. Treatment of pluripotent cells
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US10351820B2 (en) 2008-06-30 2019-07-16 Janssen Biotech, Inc. Methods for making definitive endoderm using at least GDF-8
US9593305B2 (en) 2008-06-30 2017-03-14 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9593306B2 (en) 2008-06-30 2017-03-14 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US10233421B2 (en) 2008-06-30 2019-03-19 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9012218B2 (en) 2008-10-31 2015-04-21 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9388387B2 (en) 2008-10-31 2016-07-12 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9234178B2 (en) 2008-10-31 2016-01-12 Janssen Biotech, Inc. Differentiation of human pluripotent stem cells
US9752126B2 (en) 2008-10-31 2017-09-05 Janssen Biotech, Inc. Differentiation of human pluripotent stem cells
US9969973B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Methods and compositions for cell attachment and cultivation on planar substrates
US9969972B2 (en) 2008-11-20 2018-05-15 Janssen Biotech, Inc. Pluripotent stem cell culture on micro-carriers
US10471104B2 (en) 2009-07-20 2019-11-12 Janssen Biotech, Inc. Lowering blood glucose
US8785184B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US10076544B2 (en) 2009-07-20 2018-09-18 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US8785185B2 (en) 2009-07-20 2014-07-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9593310B2 (en) 2009-12-23 2017-03-14 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9133439B2 (en) 2009-12-23 2015-09-15 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9150833B2 (en) 2009-12-23 2015-10-06 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US10704025B2 (en) 2009-12-23 2020-07-07 Janssen Biotech, Inc. Use of noggin, an ALK5 inhibitor and a protein kinase c activator to produce endocrine cells
US9969981B2 (en) 2010-03-01 2018-05-15 Janssen Biotech, Inc. Methods for purifying cells derived from pluripotent stem cells
US10329534B2 (en) 2010-03-01 2019-06-25 Janssen Biotech, Inc. Methods for purifying cells derived from pluripotent stem cells
US9752125B2 (en) 2010-05-12 2017-09-05 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9181528B2 (en) 2010-08-31 2015-11-10 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US9528090B2 (en) 2010-08-31 2016-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9951314B2 (en) 2010-08-31 2018-04-24 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9506036B2 (en) 2010-08-31 2016-11-29 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US9458430B2 (en) 2010-08-31 2016-10-04 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
US10358628B2 (en) 2011-12-22 2019-07-23 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into single hormonal insulin positive cells
US11377640B2 (en) 2011-12-22 2022-07-05 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into single hormonal insulin positive cells
US9434920B2 (en) 2012-03-07 2016-09-06 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
US9593307B2 (en) 2012-03-07 2017-03-14 Janssen Biotech, Inc. Defined media for expansion and maintenance of pluripotent stem cells
US10208288B2 (en) 2012-06-08 2019-02-19 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
US10066210B2 (en) 2012-06-08 2018-09-04 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells
US10138465B2 (en) 2012-12-31 2018-11-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using HB9 regulators
US10947511B2 (en) 2012-12-31 2021-03-16 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using thyroid hormone and/or alk5, an inhibitor of tgf-beta type 1 receptor
US10377989B2 (en) 2012-12-31 2019-08-13 Janssen Biotech, Inc. Methods for suspension cultures of human pluripotent stem cells
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
US10344264B2 (en) 2012-12-31 2019-07-09 Janssen Biotech, Inc. Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
US10006006B2 (en) 2014-05-16 2018-06-26 Janssen Biotech, Inc. Use of small molecules to enhance MAFA expression in pancreatic endocrine cells
US10870832B2 (en) 2014-05-16 2020-12-22 Janssen Biotech, Inc. Use of small molecules to enhance MAFA expression in pancreatic endocrine cells
US10420803B2 (en) 2016-04-14 2019-09-24 Janssen Biotech, Inc. Differentiation of pluripotent stem cells to intestinal midgut endoderm cells

Also Published As

Publication number Publication date
US20040072344A1 (en) 2004-04-15
JP2005514944A (en) 2005-05-26
WO2003062405A3 (en) 2003-10-16
US20030162290A1 (en) 2003-08-28

Similar Documents

Publication Publication Date Title
US20040072344A1 (en) Method for inducing differentiation of embryonic stem cells into functioning cells
US9624472B2 (en) Production of insulin producing cells
Nishimura et al. Potential use of embryonic stem cells for the treatment of mouse parkinsonian models: improved behavior by transplantation of in vitro differentiated dopaminergic neurons from embryonic stem cells
KR102487142B1 (en) How to differentiate pluripotent cells
RU2345133C2 (en) Production of terminal-differentiated dofaminnergic neurones from embrionic human founder cells
Ishii et al. In vitro differentiation and maturation of mouse embryonic stem cells into hepatocytes
US20040121460A1 (en) Differentiation of stem cells to pancreatic endocrine cells
CN1894401B (en) Terminally differentiated dopaminergic neurons derived from human embryonic stem cells
US20030003574A1 (en) Multipotent stem cells from peripheral tissues and uses thereof
KR20170064542A (en) TGFβ SIGNALING INDEPENDENT NAIVE INDUCED PLURIPOTENT STEM CELLS, METHODS OF MAKING AND USE
IL173889A (en) In vitro enriched population of gabaergic neurons for use in cell therapy
JP2021516066A (en) How to promote the differentiation of stem cells into beta cells
Lowry et al. Multipotent embryonic spinal cord stem cells expanded by endothelial factors and Shh/RA promote functional recovery after spinal cord injury
US20200277567A1 (en) Methods for chemically induced lineage reprogramming
Pokrywczynska et al. Differentiation of stem cells into insulin-producing cells: current status and challenges
H Parsons et al. Patents on technologies of human tissue and organ regeneration from pluripotent human embryonic stem cells
JP2022069552A (en) Induction of hepatocytes by stem cell differentiation with RNA
JP2024045609A (en) Induction of pancreatic β cells by stem cell differentiation with RNA
WO2018139600A1 (en) Endodermal cell mass, and method for producing any one of three primary germ layer cell mass from pluripotent cells
KR101792865B1 (en) Method for inducing differentiation of pluripotent stem cell into insulin producing cells using co-culture with mature islet cells
WO2023097513A1 (en) Method of generating functional islets from pluripotent stem cells
WO2024008810A1 (en) Differentiation of stem cells to pancreatic endocrine cells
JP2024513912A (en) Dopaminergic precursor cells (PRECURSOR CELL) and how to use them

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003562273

Country of ref document: JP

122 Ep: pct application non-entry in european phase